WO1998040509A1 - Chimeric adenoviral coat protein and methods of using same - Google Patents

Chimeric adenoviral coat protein and methods of using same Download PDF

Info

Publication number
WO1998040509A1
WO1998040509A1 PCT/US1998/005033 US9805033W WO9840509A1 WO 1998040509 A1 WO1998040509 A1 WO 1998040509A1 US 9805033 W US9805033 W US 9805033W WO 9840509 A1 WO9840509 A1 WO 9840509A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
asn
thr
leu
ala
Prior art date
Application number
PCT/US1998/005033
Other languages
French (fr)
Inventor
Ronald G. Crystal
Erik Falck-Pedersen
Jason Gall
Imre Kovesdi
Thomas J. Wickham
Original Assignee
Cornell Research Foundation, Inc.
Genvec, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cornell Research Foundation, Inc., Genvec, Inc. filed Critical Cornell Research Foundation, Inc.
Priority to CA002283628A priority Critical patent/CA2283628A1/en
Priority to JP53987998A priority patent/JP2001517080A/en
Priority to AU64652/98A priority patent/AU745252B2/en
Priority to EP98910401A priority patent/EP0973927A1/en
Publication of WO1998040509A1 publication Critical patent/WO1998040509A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10345Special targeting system for viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/40Vectors comprising a peptide as targeting moiety, e.g. a synthetic peptide, from undefined source
    • C12N2810/405Vectors comprising RGD peptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6009Vectors comprising as targeting moiety peptide derived from defined protein from viruses dsDNA viruses
    • C12N2810/6018Adenoviridae
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses

Definitions

  • the present invention relates to a chimeric adenoviral coat protein and a recombinant adenovirus comprising same.
  • the invention provides a chimeric adenoviral hexon protein and a recombinant adenovirus comprising the chimeric adenoviral hexon protein.
  • a recombinant adenovirus can be employed inter alia in gene therapy.
  • In vivo gene therapy is a strategy in which nucleic acid, usually in the form of DNA, is administered to modify the genetic repertoire of target cells for therapeutic purposes. This can be accomplished efficiently using a recombinant adenoviral vector encoding a so-called "therapeutic gene".
  • a therapeutic gene is generally considered a gene that corrects or compensates for an underlying protein deficit or, alternately, a gene that is capable of down-regulating a particular gene, or counteracting the negative effects of its encoded product, in a given disease state or syndrome.
  • Recombinant adenoviral vectors have been used to transfer one or more recombinant genes to diseased cells or tissues in need of treatment.
  • adenoviral vectors are preferred over other vectors commonly employed for gene therapy (e.g., retroviral vectors) since adenoviral vectors can be produced in high titers (i.e., up to 10 13 viral particles/ml), and they efficiently transfer genes to nonreplicating, as well as replicating, cells. Moreover, adenoviral vectors are additionally preferred based on their normal tropism for the respiratory epithelium in cases where the targeted tissue for somatic gene therapy is the lung, as well as for other reasons (see, e.g., Straus, Tn Adenoviruses, Plenan Press, New York, NY, 451-496 (1984)); Horwitz et al .
  • adenoviral serotypes There are 49 human adenoviral serotypes, categorized into 6 subgenera (A through F) based on nucleic acid comparisons, fiber protein characteristics, and biological properties (Crawford-Miksza et al . , J. Virol., 70, 1836- 1844 (1996)).
  • the group C viruses e.g., serotypes 2 and 5, or Ad2 and Ad5
  • serotypes 2 and 5 and Ad2 and Ad5 are well characterized. It is these serotypes that currently are employed for gene transfer studies, including human gene therapy trials (see, e.g., Rosenfeld et al .
  • group A e.g., serotypes 12 and 31
  • group B e.g., serotypes 3 and 7
  • group D e.g., serotypes 8 and 30
  • group E e.g., serotype 4
  • group F e.g., serotypes 40 and 41
  • Adenoviruses examined to date are nonenveloped, regular icosahedrons of about 65 to 80 nanometers in diameter.
  • Adenoviruses are comprised of linear, double-stranded DNA that is complexed with core proteins and surrounded by the adenoviral capsid.
  • the capsid is comprised of 252 capsomeres, of which 240 are hexons and 12 are pentons .
  • the hexon capsomere provides structure and form to the capsid (Pettersson, in The Adenoviruses, pp.
  • the penton comprises a penton base, which is bound to other hexon capsomeres, and a fiber, which is noncovalently bound to, and projects from, the penton base.
  • the penton fiber protein comprises three identical polypeptides (i.e., polypeptide IV).
  • the Ad2 penton base protein comprises five identical polypeptides (i.e., polypeptide III) of 571 amino acids each (Boudin et al., Virology, 92, 125-138 (1979)).
  • adenoviruses provide an elegant and efficient means of transferring therapeutic genes into cells.
  • one problem encountered with the use of adenoviral vectors for gene transfer in vivo is the generation of antibodies to antigenic epitopes on adenoviral capsid proteins. If sufficient in titer, the antibodies can limit the ability of the vector to be used more than once as an effective gene transfer vehicle.
  • adenoviral type 2 or 5 gene transfer vector can result in the production of antibodies directed against the vector which prevent expression from the same serotype vector administered 1 to 2 weeks later (see, e.g., Yei et al., supra; Zabner (1994), supra; Setoguchi et al . , Am. J. Respir. Cell. Mol. Biol., 10, 369-377 (1994); Kass-Eisler et al . , Gene Therapy, 1, 395- 402 (1994); Kass-Eisler et al . , Gene Therapy 3, 154-162 (1996)).
  • the hexon protein and to a lesser extent the fiber protein, comprise the main antigenic determinants of the virus, and also determine the serotype specificity of the virus (Watson et al . , J. Gen. Virol., 69, 525-535 (1988); Wolfort et al., J. Virol., 62:, 2321-2328 (1988); Wolfort et al., J. Virol., 5_6, 896-903 (1985); Crawford-Miksza et al . , supra) .
  • researchers have examined and compared the structure of these coat proteins of different adenoviral serotypes in an effort to define the regions of the proteins against which neutralizing antibodies are elicited.
  • the Ad2 hexon trimer is comprised of a pseudohexagonal base and a triangular top formed of three towers (Roberts et al . , supra; Athappilly et al . , J. Mol. Biol. , 242, 430-455 (1994)).
  • the base pedestal consists of two tightly packed eight-stranded antiparallel beta barrels stabilized by an internal loop.
  • the predominant regions in hexon protein against which neutralizing antibodies are directed appear to be in loops 1 and 2 (i.e., LI or 11, and LII or 12 , respectively) in one of the three towers.
  • Kloch et al J. Biol. Chem.
  • hexon protein is composed of seven discrete hypervariable regions in loops and 1 and 2 (HVRl to HVR7 ) which vary in length and sequence between adenoviral serotypes (Crawford-Miksza et al., supra) .
  • the trimeric fiber protein consists of a tail, a shaft, and a knob (Devaux et al., J. Molec. Biol., 215, 567-588 (1990)).
  • the fiber shaft region is comprised of repeating 15 amino acid motifs, which are believed to form two alternating beta strands and beta bends (Green et al . , EMBO J . , 2_, 1357- 1365 (1983)).
  • the overall length of the fiber shaft region and the number of 15 amino acid repeats differ between adenoviral serotypes.
  • the receptor binding domain of the fiber protein and sequences necessary for fiber trimerization are localized in the knob region encoded by roughly the last 200 amino acids of the protein (Henry et al., J. Virol., 68 (8) , 5239-5246 (1994)); Xia et al., Structure, 2(12), 1259-1270 (1994)). Furthermore, all adenovirus serotypes appear to possess a type of specific moiety located in the knob region (Toogood et al . , supra . )
  • adenovirus as a vector for gene therapy. Accordingly, recombinant adenoviral vectors capable of escaping such neutralizing antibodies (in the event they are preexisting and hamper gene expression commanded by adenovirus in an initial dose) , and which would allow repeat doses of adenoviral vectors to be administered, would significantly advance current gene therapy methodology.
  • the present invention seeks to overcome at least some of the aforesaid problems of recombinant adenoviral gene therapy.
  • the present invention provides a chimeric adenovirus coat protein (particularly a chimeric adenovirus hexon protein) comprising a nonnative amino acid sequence.
  • the chimeric adenovirus coat protein is not recognized by, or has a decreased ability to be recognized by, a neutralizing antibody directed against the corresponding wild-type (i.e., native) coat protein.
  • the chimeric adenovirus coat protein enables a vector (such as an adenovirus) comprising the corresponding protein to be administered repetitively, or to be administered following administration of an adenovirus vector comprising the corresponding wild-type coat protein.
  • a vector such as an adenovirus
  • the present invention also provides a vector, particularly an adenoviral vector, that comprises a chimeric adenovirus coat protein such as chimeric adenovirus hexon protein (and which optionally further comprises a chimeric adenovirus fiber and/or penton base protein) , and methods of constructing and using such a vector .
  • Figure 1 is a diagram of the method employed to construct the vector pAd70-100dlE3. fiber7.
  • Figure 2 is a partial restriction map of the vector pGBS.59-100(HSF:RGD) .
  • the present invention provides, among other things, a chimeric adenovirus coat protein.
  • the chimeric adenovirus coat protein comprises a nonnative amino acid sequence, such that the chimeric adenovirus coat protein (or a vector comprising the chimeric adenovirus coat protein) has a decreased ability or inability to be recognized by antibodies (e.g., neutralizing antibodies) directed against the corresponding wild-type adenovirus coat protein .
  • a "coat protein" according to the invention is either an adenoviral penton base protein, an adenoviral hexon protein, or an adenoviral fiber protein.
  • a coat protein is a adenoviral hexon protein or an adenoviral fiber protein. Any one of the serotypes of human or nonhuman adenovirus can be used as the source of the coat protein, or its gene or coding sequence.
  • the adenovirus coat protein is that of a Group B or C adenovirus and, preferably, is that of Adl, Ad2, Ad3, Ad5, Ad6, Ad7 , Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, or Ad48.
  • the chimeric adenovirus coat protein (or a vector, such as adenoviral vector, comprising the chimeric adenovirus coat protein) has a decreased ability or an inability to be recognized by an antibody (e.g., a neutralizing antibody) directed against the corresponding wild-type adenovirus coat protein.
  • an antibody e.g., a neutralizing antibody
  • a "neutralizing antibody” is an antibody that either is purified from or is present in serum.
  • an antibody can be a single antibody or a plurality of antibodies.
  • An antibody is "neutralizing” if it inhibits infectivity of (i.e., cell entry) or gene expression commanded by an adenovirus comprising wild-type coat protein, or if it exerts a substantial deleterious effect on infectivity of or gene expression commanded by an adenovirus comprising wild-type coat protein, as compared, for instance, to any effect on any other adenoviral property.
  • An ability or inability of a chimeric coat protein to "be recognized by” (i.e., interact with) a neutralizing antibody directed against the wild-type adenovirus coat protein can be assessed by a variety of means known to those skilled in the art. For instance, the removal of one or more epitopes for a neutralizing antibody present in a wild-type adenovirus coat protein to generate a chimeric adenovirus coat protein will result in a decreased ability or inability of the chimeric coat protein to be recognized by the neutralizing antibody. Also, such a decreased ability or inability to interact with a neutralizing antibody directed against wild-type coat protein can be demonstrated by means of a neutralization test (see, e.g., Toogood et al . , supra; Crawford-Miksza et al . , supra; Mastrangeli et al., Human Gene Therapy, 1_, 79-87 (1996) ) , or as further described herein .
  • an "inability" of a chimeric adenovirus coat protein (or a vector comprising a chimeric adenovirus coat protein) to be recognized by a neutralizing antibody directed against wild-type adenovirus coat protein means that such an antibody does not interact with the chimeric coat protein, and/or exhibits no substantial deleterious effect on infectivity of or gene expression commanded by an adenovirus comprising wild-type coat protein, as compared, for instance, to any effect on any other adenoviral property.
  • a "decreased ability" to be recognized by neutralizing antibody directed against wild-type adenovirus coat protein refers to any decrease in the ability of the chimeric adenovirus coat protein (or a vector comprising the chimeric coat protein) to be recognized by an antibody directed against the corresponding wild-type adenovirus coat protein as compared to the wild-type adenovirus coat protein.
  • a "decreased ability" to be recognized by a neutralizing antibody directed against wild-type adenovirus coat protein is exhibited by from about a 10% to about a 99% increase in the ability of a recombinant adenovirus comprising the chimeric coat protein to cause a visible cytopathic effect (c.p.e.) in cells such as A549 cells or COS-1 cells (or other such cells appropriate for a neutralization assay) in the presence of the neutralizing antibody as compared to an adenovirus comprising the wild-type coat protein against which the neutralizing antibody is directed.
  • a visible cytopathic effect c.p.e.
  • a decreased ability or inability of an adenovirus chimeric coat protein (or a vector comprising a chimeric adenovirus coat protein) to interact with a neutralizing antibody can be shown by a reduction of inhibition (from about 10% to about 99%) or no inhibition at all of cell infectivity by a recombinant vector (such as an adenoviral vector) containing the chimeric coat protein as compared to a recombinant vector containing the wild-type protein.
  • a recombinant vector such as an adenoviral vector
  • a decreased ability or inability of an adenovirus chimeric coat protein (or a vector comprising a chimeric adenovirus coat protein) to interact with a neutralizing antibody can be shown by a reduction of inhibition (from about 10% to about 99%) or no inhibition at all of gene expression commanded by a recombinant vector (such as an adenoviral vector) containing the chimeric coat protein as compared to a recombinant vector containing the wild-type coat protein.
  • a recombinant vector such as an adenoviral vector
  • the coat protein is "chimeric” in that it comprises a sequence of amino acid residues that is not typically found in the protein as isolated from, or identified in, wild-type adenovirus, which comprises the so-called native coat protein, or "wild-type coat protein” .
  • the chimeric coat protein thus comprises (or has) a "nonnative amino acid sequence".
  • “nonnative amino acid sequence” is meant any amino acid sequence (i.e., either component residues or order thereof) that is not found in the native coat protein of a given serotype of adenovirus, and which preferably is introduced into the coat protein at the level of gene expression (i.e., by production of a nucleic acid sequence that encodes the nonnative amino acid sequence) .
  • the nonnative amino acid sequence can be obtained by deleting a portion of the amino acid sequence, deleting a portion of the amino acid sequence and replacing the deleted amino sequence with a so-called "spacer region", or introducing the spacer region into an unmodified coat protein.
  • spacer region a so-called "spacer region"
  • such manipulations result in a chimeric adenovirus coat protein according to the invention that is capable of carrying out the functions of the corresponding wild-type adenovirus coat protein (or, at least that when incorporated into an adenovirus, will allow appropriate virion formation and will not preclude adenoviral-mediated cell entry) , and, optimally, that is not impeded in its proper folding.
  • a nonnative amino acid sequence according to the invention preferably comprises a deletion of a region of a wild-type adenovirus coat protein, particularly an adenovirus hexon or fiber protein.
  • the resultant nonnative amino acid sequence is such that one or more of the existing epitopes for neutralizing antibodies directed against the corresponding wild-type adenovirus coat protein have been rendered non- immunogenic.
  • the region deleted comprises from about 1 to about 750 amino acids, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted comprises a smaller region less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids.
  • the chimeric coat protein also desirably comprises a plurality of such deletions.
  • the chimeric adenovirus coat protein comprises modification of one or more amino acids, and such modification is made in one or more regions .
  • a nonnative amino acid sequence comprises a deletion of one or more regions of a wild-type adenovirus hexon protein, wherein preferably the hexon protein is the Ad2 hexon protein [SEQ ID NO: 2] (which is encoded by the sequence of SEQ ID NO:l; GenBank® Data Bank Accession Number U20821), or the Ad5 hexon protein [SEQ ID NO: 3] (GenBank® Data Bank Accession Number M73260, which is encoded by the sequence of SEQ ID NO:4), or the Ad7 hexon protein (GenBank® Data Bank Accession Number x76551) .
  • the hexon protein is the protein sequence reported by Crawford-Miksza et al . (Ad2 hexon [SEQ ID NO:52], Ad5 hexon SEQ ID NO:54]) .
  • the sequences of Crawford-Miksza et al . differ over those reported in the GenBank® Data Bank in that the amino acid residue reported as the first in the Crawford-Miksza et al . sequences is not Met, and the Ad5 hexon sequence is reported as terminating with "Gin His" instead of with "Thr Thr” .
  • the numbering of adenovirus hexon amino acid residues corresponds to that in Crawford-Miksza et al.
  • the region (s) of the deletion comprises an internal hexon protein sequence ("internal” meaning not at or near the C- or N-terminus of the protein; "near” referring to a distance of 500 amino acids or less ) , preferably a hypervariable region, e.g., as reported in Crawford-Miksza et al .
  • the internal region of the wild-type hexon protein that is deleted to generate the chimeric hexon protein comprises the entirety of 11 loop, preferably from about residue 131 to about residue 331 of the Ad2 hexon protein [SEQ ID NO: 6] (which is encoded by the sequence of SEQ ID NO: 5), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad5 [SEQ ID NO: 8] (which is encoded by the sequence of SEQ ID NO:7), Ad6, Ad7 , Ad8, Adl2, Adl ⁇ , Ad40, Ad41, Ad48, BAV3, or MAV1, especially as reported in Crawford- Miksza et al . , supra .
  • the internal region of the wild-type hexon protein that is deleted to produce the chimeric hexon protein comprises one or more regions (e.g., smaller regions) of the 11 loop.
  • the region deleted comprises a hypervariable region.
  • the one or more regions of the II loop deleted are regions (i.e., hypervariable regions) selected from this group consisting of the HVRl region, the HVR2 region, the HVR3 region, the HVR4 region, the HVR5 region, and the HVR6 region.
  • the region of the wild- type protein that is deleted occurs on the external surface of the hexon protein.
  • HVR2, HVR3, HVR4 , and HVR5 each of which are externally located regions of the hexon protein -- are particularly preferred for deletion or modification .
  • the "HVRl region” preferably comprises from about amino acid 137 to about amino acid 188 of the Ad2 hexon protein [SEQ ID NO: 10] (which is encoded by the sequence of SEQ ID NO: 9), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 12] (which is encoded by the sequence of SEQ ID NO:ll), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the "HVR2 region” preferably comprises from about amino acid 194 to about amino acid 204 of the Ad2 hexon protein [SEQ ID NO: 14] (which is encoded by the sequence of SEQ ID NO: 13), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 16] (which is encoded by the sequence of SEQ ID NO:15), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the "HVR3 region” preferably comprises from about amino acid 222 to about amino acid 229 of the Ad2 hexon protein [SEQ ID NO: 18] (which is encoded by the sequence of SEQ ID NO:17), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO:20] (which is encoded by the sequence of SEQ ID NO:19), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl ⁇ , Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the "HVR4 region” preferably comprises from about amino acid 258 to about amino acid 271 of the Ad2 hexon protein [SEQ ID NO: 22] (which is encoded by the sequence of SEQ ID NO:21), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 24] (which is encoded by the sequence of SEQ ID NO:23), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the "HVR5 region” preferably comprises from about amino acid 278 to about amino acid 294 of the Ad2 hexon protein [SEQ ID NO:26] (which is encoded by the sequence of SEQ ID NO:25), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO:28] (which is encoded by the sequence of SEQ ID NO:27), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the deleted region comprises from about amino acid 297 to about amino acid 304 just outside of the HVR5 region of the Ad2 hexon protein [SEQ ID NO: 30] (which is encoded by the sequence of SEQ ID NO:29), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 32] (which is encoded by the sequence of SEQ ID NO:31), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the "HVR6 region” preferably comprises from about amino acid 316 to about amino acid 327 of the Ad2 hexon protein [SEQ ID NO: 34] (which is encoded by the sequence of SEQ ID NO:33), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 36] (which is encoded by the sequence of SEQ ID NO:35), Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the internal region of the wild-type hexon protein that is deleted to generate the chimeric hexon protein comprises the entirety of the 12 loop, preferably from about residue 423 to about residue 477 of the Ad2 hexon protein [SEQ ID NO:38] (which is encoded by the sequence of SEQ ID NO:37), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 40] (which is encoded by the sequence of SEQ ID NO:39), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al .
  • the internal region of the wild-type hexon protein that is deleted to produce the chimeric hexon protein comprises one or more smaller regions (e.g., hypervariable regions) of the 12 loop.
  • the smaller region of the 12 loop comprises the HVR7 region.
  • the "HVR7 region” preferably comprises from about amino acid 433 to about amino acid 465 of the Ad2 hexon protein [SEQ ID NO: 42] (which is encoded by the sequence of SEQ ID NO:41), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 44] (which is encoded by the sequence of SEQ ID NO:43), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
  • the deleted region comprises from about amino acid 460 to about amino acid 466 of the HVR7 region (i.e., extending one base pair outside of this region) of the Ad2 hexon protein [SEQ ID NO:46] (which is encoded by the sequence of SEQ ID NO:45), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 48] (which is encoded by the sequence of SEQ ID NO:47), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al., supra .
  • the chimeric adenovirus hexon protein desirably comprises deletions in one or both of the aforementioned regions, i.e., the hexon protein comprises deletions in one or both of the 11 and 12 loops, which deletions can constitute the entirety of the loop(s), or can comprise deletions of one or more smaller regions (e.g., hypervariable regions) in one or both of the hexon loops.
  • the deleted region (s) are selected from the group consisting of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO: 46, and SEQ ID NO: 48, and equivalents and conservative variations of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO
  • an “equivalent” is a naturally occurring variation of an amino acid or nucleic acid sequence, e.g., as are observed among different strains of adenovirus.
  • a conservative variation is a variation of an amino acid sequence that results in one or more conservative amino acid substitution (s) .
  • a “conservative amino acid substitution” is an amino acid substituted by an alternative amino acid of similar charge density, hydrophilicity/hydrophobicity, size, and/or configuration
  • a chimeric adenoviral fiber or hexon protein comprises a deletion of one or more region (s) of the wild-type adenovirus coat protein (particularly the 11 and/or 12 loops, and, most particularly, the HVRl, HVR2, HVR3, HVR4, HVR5, HVR6, and/or HVR7 regions of the wild- type adenovirus hexon protein) as previously described, and further comprises a replacement of the region (s) with a spacer region preferably of from 1 to about 750 amino acids, especially of from about 1 to about 500 amino acids, and particularly of from about 1 to about 300 amino acids.
  • the region deleted and replaced comprises a smaller region less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids.
  • the chimeric coat protein also desirably comprises a plurality of such replacements.
  • the chimeric adenovirus coat protein comprises modification of one or more amino acids, and such modification is made in one or more regions which can be a smaller region.
  • a spacer region of the aforementioned size also preferably simply can be inserted into one of the aforementioned regions (particularly into the 11 and/or 12 loop, or one or more of the aforementioned HVRl, HVR2, HVR3, HVR4 , HVR5, HVR6, and HVR7 regions of the adenovirus hexon protein) in the absence of any deletion to render the resultant chimeric protein nonimmunogenic by, for instance, destroying the ability of a neutralizing antibody to interact with that particular site (e.g., by changing the spatial juxtaposition of critical amino acids with which the antibody interacts) .
  • the spacer region comprises a nonconservative variation of the amino acid sequence of wild-type adenovirus coat protein (particularly wild-type adenovirus hexon protein) that comprises an epitope for a neutralizing antibody, and which may or may not be deleted upon the insertion of the spacer region.
  • a "nonconservative variation” is a variation of this amino acid sequence that does not result in the creation or recreation in the chimeric adenovirus coat protein of the epitope for a neutralizing antibody directed against the wild-type adenovirus coat protein, and, in particular, is a variation of the spacer region that results in one or more nonconservative amino acid insertion (s) or substitution (s) in this region.
  • nonconservative amino acid substitution is an amino acid substituted by an alternative amino acid of differing charge density, hydrophilicity/hydrophobicity, size, and/or configuration (e.g., a change of a basic amino acid for an acidic amino acid, a hydrophilic amino acid for a hydrophobic amino acid, and the like) .
  • the spacer region does not interfere with the functionality of the chimeric adenovirus coat protein, particularly the chimeric adenovirus hexon or fiber protein, e.g., the ability of hexon protein to bind penton base protein or other hexon capsomeres, or the ability of penton fiber to bind penton base and/or to a cell surface receptor.
  • functionality can be assessed by virus viability.
  • the absence of the creation or recreation of the epitope (s) for a neutralizing antibody directed against the wild-type coat (e.g., hexon and/or fiber) protein can be confirmed using techniques as described in the Examples which follow (e.g., by ensuring the antibody, which may be in a carrier fluid such as . serum or other liquid, binds the wild-type adenovirus coat protein, but not the chimeric adenovirus coat protein) .
  • the spacer region incorporated into the adenovirus coat protein comprises a series of polar and/or charged amino acids (e.g., Lys, Arg, His, Glu, Asp, and the like), or amino acids with intermediate polarity (e.g., Gin, Asn, Thr, Ser, Met, and the like) .
  • the spacer region comprises the sequence of SEQ ID NO: 50 (which is encoded by the sequence of SEQ ID NO:49), and equivalents and conservative variations of SEQ ID NO: 50.
  • the spacer region can comprise any other sequence like the FLAG octapeptide sequence of SEQ ID NO: 50 that will not interfere with the functionality of the resultant chimeric protein.
  • a region of a wild-type adenovirus coat protein is deleted and replaced with a spacer region comprising the corresponding coat protein region of another adenoviral serotype.
  • the spacer region is of a different adenoviral group.
  • a region of an Ad2 coat protein can be replaced with the corresponding region of an Ad5 or Ad7 coat protein (or any other serotype of adenovirus as described above) , and vice versa.
  • such a spacer region comprising the coat protein region of another adenoviral serotype is simply inserted into the corresponding coat protein region of the chimeric coat protein.
  • the likelihood of obtaining a chimeric hexon protein that is functional can be increased by making sure that the size of the hypervariable domain resulting from such insertion approximates the size of a known hypervariable domain.
  • the HVRl region of Ad40 is about 30 amino acids smaller than the HVRl region of Ad2 (as well as other adenoviruses such as Ad5, Ad8, etc.) .
  • a spacer region of about 30 amino acids can be incorporated into the Ad40 HVRl region to produce a chimeric adenovirus hexon protein.
  • the region of Ad2 (or other adenovirus) that is not present in Ad40 i.e., approximately amino acid residues 138 to 174, or a portion thereof, is introduced into Ad40 to produce the chimeric adenoviral hexon protein .
  • the nonnative amino acid sequence of a chimeric coat protein comprises a plurality of such replacements or insertions.
  • the coat protein is incorporated into an adenoviral vector, preferably the entire coat protein of one adenoviral serotype can be substituted with the entire coat protein of another adenoviral serotype, as described further herein .
  • the region or regions of wild-type adenovirus hexon protein that are deleted and replaced by the spacer region, or into which the spacer region is inserted can be any suitable region (s) and desirably comprise one or more of the regions described above with respect to the hexon protein deletions.
  • the one or more regions into which the spacer region is inserted or which the spacer region replaces comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48, and equivalents and conservative variations of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:
  • the spacer region itself preferably comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID N0:12, SEQ ID N0:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48, and equivalents and conservative variations of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:
  • the fiber protein also preferably is altered in a similar fashion as described for modification of hexon protein to escape antibodies directed in particular against wild-type adenovirus fiber protein.
  • Fiber protein sequences and methods of modifying fiber protein are known to those skilled in the art (see, e.g., Xia et al . , supra; Novelli et al . , Virology, 185, 365-376 (1991)) .
  • the fiber manipulations can be carried out in the absence of, or along with, modifications to the adenovirus hexon protein.
  • the fiber protein can be replaced in its entirety, or in part, with sequences of a fiber protein from a different serotype of adenovirus.
  • deletions can be made of fiber sites that constitute an epitope for a neutralizing antibody, and/or insertions can be made at the site to destroy the ability of the protein to interact with the antibody.
  • the chimeric adenovirus coat protein (particularly the chimeric adenovirus hexon or fiber protein) comprises a nonnative amino acid sequence wherein the alteration is made at the level of DNA.
  • the invention preferably provides an isolated and purified nucleic acid encoding a chimeric adenovirus coat protein.
  • the invention provides an isolated and purified nucleic acid encoding a chimeric adenovirus hexon protein as defined herein, wherein the nucleic acid sequence comprises a deletion of a region (or a plurality of such deletions) that encodes from about 1 to about 750 amino acids of the wild-type adenovirus coat protein, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted comprises a smaller region that encodes less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids.
  • the deletion comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO:47, or a sequence comprising the corresponding region from Adl, Ad3, Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48,
  • the invention also preferably provides an isolated and purified nucleic acid encoding a chimeric adenovirus hexon protein as defined herein, wherein the nucleic acid sequence comprises a deletion of one or more sequences selected from the group consisting of equivalents and conservatively modified variants of sequences that encode the entirety of the II and/or 12 loop, or SEQ ID NO: 5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID N0:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID N0:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO: 7, or a sequence comprising the corresponding region from
  • an “equivalent” is a variation on the nucleic acid sequence such as can occur in different strains of adenovirus, and which either does or does not result in a variation at the amino acid level. Failure to result in variation at the amino acid level can be due, for instance, to degeneracy in the triplet code.
  • a “conservatively modified variant” is a variation on the nucleic acid sequence that results in one or more conservative amino acid substitutions .
  • a “nonconservatively modified variant” is a variation on the nucleic acid sequence that results in one or more nonconservative amino acid substitutions .
  • the invention provides an isolated and purified nucleic acid encoding a chimeric adenovirus coat protein wherein the nucleic acid sequence further comprises a replacement of the deleted region (or a plurality of such replacements) with a spacer nucleic acid region (i.e., the nucleic acid sequence that encodes the aforementioned "spacer region") that encodes from about 1 to about 750 amino acids of the wild-type adenovirus coat protein, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted and replaced comprises a smaller region that encodes less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids.
  • the spacer nucleic acid region comprises a FLAG octapeptide-encoding sequence [SEQ ID NO:49], and equivalents and conservatively modified variants of SEQ ID NO: 49.
  • a spacer nucleic acid region can be employed that substitutes one or more coat protein encoding regions (particularly a hexon protein encoding region) of a particular adenoviral serotype with a coat protein encoding region (particularly a hexon protein encoding region) of another adenoviral serotype.
  • a spacer nucleic acid region present in a chimeric adenoviral hexon protein is selected from the group consisting of sequences that encode the entirety of the 11 and/or 12 loop, or SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO:47, or a sequence comprising the corresponding region from Adl, Ad3, Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40,
  • the spacer nucleic acid region (or a plurality thereof) simply can be incorporated into the coat protein in the absence of any deletions . These manipulations can be carried out so as to produce the above-described chimeric adenovirus coat protein.
  • chimeric adenoviral coat protein i.e., by introducing conservative or nonconservative variations at either the level of DNA or protein
  • methods of making such a chimeric adenoviral coat protein are known in the art, are described in the Examples which follow, and also can be accomplished by means of various commercially available kits and vectors (e.g., New England Biolabs, Inc., Beverly, MA; Clontech, Palo Alto, CA; Stratagene, LaJolla, CA, and the like) .
  • the ExSiteTM PCR-based site-directed mutagenesis kit and the ChameleonTM double-stranded site- directed mutagenesis kit by Stratagene can be employed for introducing such mutations.
  • the means of assessing such mutations are described in the Examples herein.
  • the present invention provides a preferred means of making a chimeric adenoviral coat protein, particularly a chimeric adenoviral hexon protein, which comprises obtaining an adenoviral genome encoding the wild-type adenovirus coat protein (e.g., the wild-type adenovirus hexon protein) , and deleting one or more region (s) of the chimeric adenovirus coat protein (particularly the chimeric adenovirus hexon protein) comprising from about 1 to about 750 amino acids by modifying the corresponding nucleic acid coding sequence.
  • the wild-type adenovirus coat protein e.g., the wild-type adenovirus hexon protein
  • region (s) of the chimeric adenovirus coat protein particularly the chimeric adenovirus hexon protein
  • the invention provides a method of making a chimeric adenovirus coat protein (particularly a chimeric adenovirus hexon protein) which comprises obtaining an adenoviral genome encoding the wild-type adenovirus coat protein, deleting one or more region (s) of the adenovirus coat protein comprising from about 1 to about 750 amino acids by modifying the corresponding coding sequence, and replacing the deleted region (s) with a spacer region comprising from about 1 to about 300 amino acids by introducing a nucleic acid region (i.e., a "spacer nucleic acid region") that codes for same.
  • a nucleic acid region i.e., a "spacer nucleic acid region
  • the spacer region preferably is simply incorporated into the coat protein (particularly the hexon protein) in the absence of any deletion.
  • the spacer nucleic acid region encodes a nonconservative variation of the amino acid sequence of the wild-type adenovirus coat protein.
  • the size of the DNA used to replace the native coat protein coding sequence may be constrained, for example, by impeded folding of the coat protein or improper assembly of the coat protein into a complex (e.g., penton base/hexon complex) or virion.
  • DNA encoding 150 amino acids or less is particularly preferred for insertion/replacement in the chimeric coat protein gene sequence, and DNA encoding 50 amino acids or less is even more preferred.
  • the method of mutagenesis comprises deleting one or more regions of an adenovirus coat protein, and/or inserting into an adenovirus coat protein one or more regions with a differing amino acid sequence, particularly by manipulating the DNA sequence.
  • Several methods are available for carrying out such manipulations of adenovirus coat protein DNA sequences; these methods further can be used in combination. The method of choice depends on factors known to those skilled in the art, e.g., the size of the DNA region to be manipulated. For instance, convenient restriction sites (which further can be introduced into a sequence) can be used to introduce or remove segments of DNA, or entire genes or coding sequences.
  • mutant strand can be separated from the parental strand using various selection means known to those skilled in the art (see, e.g., Kunkel et al . , Methods Enzymol.
  • the parental strand can be selectively degraded, for instance, with use of enzymes that nick the nonmethylated strand of a hemi-methylated DNA molecule (e.g., Jfpall, Mspl , and Sau3AI), and by extending the mutant strand using 5-methyl-dCTP, which renders the strand resistant to cleavage by these enzymes.
  • enzymes that nick the nonmethylated strand of a hemi-methylated DNA molecule e.g., Jfpall, Mspl , and Sau3AI
  • an entirely PCR-based approach can be employed for making mutations (e.g., Kunkel, Proc. Natl . Acad. Sci., 82, 488- 492 (1985); Costa et al .
  • nucleic Acids Res. 22_, 2423 (1994)
  • amino acid substitutions or deletions can be introduced during PCR by incorporating appropriate mismatches in one or both primers.
  • the nucleic acid fragment encoding the sequence further can be isolated, e.g., by PCR amplification using 5' and 3' primers, or through use of convenient restriction sites.
  • a “vector” according to the invention is a vehicle for gene transfer as that term is understood by those skilled in the art, and includes viruses, plasmids, and the like.
  • a preferred vector is an adenovirus, particularly a virus of the family Adenoviridae, and desirably of the genus Mastadenovirus (e.g., comprised of mammalian adenoviruses) or Aviadenovirus (e.g., comprised of avian adenoviruses) .
  • Such an adenovirus can be transferred by its own means of effecting cell entry (e.g., by receptor-mediated endocytosis) , or can be transferred to a cell like a plasmid, i.e., in the form of its nucleic acid, for instance, by using liposomes to transfer the nucleic acid, or by microinjecting or transforming the DNA into the cell.
  • the nucleic acid vectors that can be employed for gene transfer, particularly the adenoviral nucleic acid vectors are referred to herein as "transfer vectors".
  • Such nucleic acid vectors also include intermediary plasmid vectors that are employed, e.g., in the construction of adenoviral vectors.
  • an adenoviral vector is a serotype group C virus, preferably an Ad2 or Ad5 vector, although any other serotype adenoviral vector (e.g., group A including serotypes 12 and 31, group B including serotypes 3 and 7, group D including serotypes 8 and 30, group E including serotype 4, and group F including serotypes 40 and 41, and other Ad vectors previously described) can be employed.
  • An adenoviral vector employed for gene transfer can be replication competent.
  • an adenoviral vector can comprise genetic material with at least one modification therein, which renders the virus replication deficient.
  • the modification to the adenoviral genome can include, but is not limited to, addition of a DNA segment, rearrangement of a DNA segment, deletion of a DNA segment, replacement of a DNA segment, or introduction of a DNA lesion.
  • a DNA segment can be as small as one nucleotide and as large as 36 kilobase pairs (i.e., the approximate size of the adenoviral genome) or, alternately, can equal the maximum amount which can be packaged into an adenoviral virion (i.e., about 38 kb) .
  • Preferred modifications to the group C adenoviral genome include modifications in the El, E2, E3 and/or E4 regions.
  • an adenoviral vector can be a cointegrate, i.e., a ligation of adenoviral sequences with other sequences, such as other virus sequences, particularly baculovirus sequences, or plasmid sequences, e.g., so as to comprise a prokaryotic or eukaryotic expression vector.
  • an adenoviral vector in terms of an adenoviral vector (particularly a replication deficient adenoviral vector) , such a vector can comprise either complete capsids (i.e., including a viral genome such as an adenoviral genome) or empty capsids (i.e., in which a viral genome is lacking, or is degraded, e.g., by physical or chemical means) .
  • the capsid further can comprise nucleic acid linked to the surface by means known in the art (e.g., Curiel et al .
  • a vector i.e., a transfer vector
  • DNA DNA
  • a transfer vector similarly can comprise DNA, in the absence of any associated protein such as capsid protein, and in the absence of any envelope lipid.
  • RNA viruses also can be employed as vectors or transfer vectors
  • a transfer vector also can comprise RNA.
  • a transfer vector typically encodes a chimeric adenoviral coat protein (particularly a chimeric adenoviral hexon and/or fiber protein) .
  • the invention provides an adenoviral vector that comprises a chimeric coat protein (particularly a chimeric hexon and/or fiber protein) according to the invention.
  • a vector comprises a chimeric coat protein (particularly a chimeric adenovirus hexon protein and/or chimeric adenovirus fiber protein) as described above.
  • the vector lacks wild-type fiber protein, e.g., the vector encodes a truncated or non-functional fiber protein, or fails to translate fiber protein.
  • fiber mutations and the means of introducing fiber mutations are known to those skilled in the art (see, e.g., Falgout et al . , J. Virol., 6_, 622-625 (1988)).
  • the chimeric adenoviral coat proteins include coat proteins in which the native (i.e., wild- type) hexon and/or fiber protein of an adenoviral vector is replaced by a hexon and or fiber amino acid sequence of a different adenoviral serotype such that the resultant adenoviral vector has a decreased ability or inability to be recognized by neutralizing antibodies directed against the corresponding wild-type coat protein.
  • This replacement can comprise the entirety of the hexon and/or fiber amino acid sequence, or only a portion, as described above.
  • Both proteins can be manipulated (e.g., in a single adenovirus), or only a single chimeric adenovirus coat protein can be employed, with the remaining coat proteins being wild-type.
  • a vector according to the invention preferably comprises additional sequences and mutations, e.g., some that can occur within the coat protein coding sequence itself.
  • a vector according to the invention further preferably comprises a nucleic acid encoding a passenger gene or passenger coding sequence.
  • a "nucleic acid” is a polynucleotide (i.e., DNA or RNA) .
  • a “gene” is any nucleic acid sequence coding for a protein or an RNA molecule. Whereas a gene comprises coding sequences plus any non-coding sequences, a "coding sequence" does not include any non-coding (e.g., regulatory) DNA.
  • a “passenger gene” or “passenger coding sequence” is any gene which is not typically present in and is subcloned into a vector (e.g., a transfer vector) according to the present invention, and which upon introduction into a host cell is accompanied by a discernible change in the intracellular environment (e.g., by an increased level of deoxyribonucleic acid (DNA) , ribonucleic acid (RNA) , peptide or protein, or by an altered rate of production or degradation thereof) .
  • a vector e.g., a transfer vector
  • a “gene product” is either an as yet untranslated RNA molecule transcribed from a given gene or coding sequence (e.g., mRNA or antisense RNA) or the polypeptide chain (i.e., protein or peptide) translated from the mRNA molecule transcribed from the given gene or coding sequence.
  • a gene or coding sequence is "recombinant” if the sequence of bases along the molecule has been altered from the sequence in which the gene or coding sequence is typically found in nature, or if the sequence of bases is not typically found in nature.
  • a gene or coding sequence can be naturally occurring or wholly or partially synthetically made, can comprise genomic or complementary DNA (cDNA) sequences, and can be provided in the form of either DNA or RNA.
  • Non-coding sequences or regulatory sequences include promoter sequences.
  • a “promoter” is a DNA sequence that directs the binding of RNA polymerase and thereby promotes RNA synthesis.
  • “Enhancers” are cis-acting elements of DNA that stimulate or inhibit transcription of adjacent genes.
  • An enhancer that inhibits transcription is also termed a “silencer”.
  • Enhancers differ from DNA-binding sites for sequence-specific DNA binding proteins found only in the promoter (which also are termed “promoter elements”) in that enhancers can function in either orientation, and over distances of up to several kilobase pairs, even from a position downstream of a transcribed region.
  • a coding sequence is "operably linked" to a promoter (e.g., when both the coding sequence and the promoter constitute a passenger gene) when the promoter is capable of directing transcription of that coding sequence .
  • a “passenger gene” can be any gene, and desirably either is a therapeutic gene or a reporter gene.
  • a passenger gene is capable of being expressed in a cell in which the vector has been internalized.
  • the passenger gene can comprise a reporter gene, or a nucleic acid sequence which encodes a protein that can be detected in a cell in some fashion.
  • the passenger gene also can comprise a therapeutic gene, for instance, a therapeutic gene which exerts its effect at the level of RNA or protein.
  • a protein encoded by a transferred therapeutic gene can be employed in the treatment of an inherited disease, such as, e.g., the cystic fibrosis transmembrane conductance regulator cDNA for the treatment of cystic fibrosis.
  • the protein encoded by the therapeutic gene can exert its therapeutic effect by resulting in cell killing.
  • expression of the gene in itself may lead to cell killing, as with expression of the diphtheria toxin A gene, or the expression of the gene may render cells selectively sensitive to the killing action of certain drugs, e.g., expression of the HSV thymidine kinase gene renders cells sensitive to antiviral compounds including acyclovir, gancyclovir and FIAU ( 1- (2-deoxy-2-fluoro-b-D- arabinofuranosil) -5-iodouracil) .
  • acyclovir, gancyclovir and FIAU 1- (2-deoxy-2-fluoro-b-D- arabinofuranosil) -5-iodouracil
  • the therapeutic gene can exert its effect at the level of RNA, for instance, by encoding an antisense message or ribozyme, by affecting splicing or 3' processing (e.g., polyadenylation) , or by encoding a protein which acts by affecting the level of expression of another gene within the cell (i.e., where gene expression is broadly considered to include all steps from initiation of transcription through production of a processed protein) , perhaps, among other things, by mediating an altered rate of mRNA accumulation, an alteration of mRNA transport, and/or a change in post-transcriptional regulation.
  • adenovirus can be used for gene therapy or to study the effects of expression of the gene (e.g., a reporter gene) in a given cell or tissue in vi tro or in vivo, or for diagnostic purposes.
  • a passenger coding sequence can be employed in the vector.
  • a coding sequence can be employed for a variety of purposes even though a functional gene product may not be translated from the vector sequence.
  • the coding sequence can be used as a substrate for a recombination reaction, e.g., to recombine the sequence with the host cell genome or a vector resident in the cell.
  • the coding sequence also can be an "anticoding sequence," e.g., as appropriate for antisense approaches. Other means of using the coding sequence will be known to one skilled in the art.
  • the present invention thus provides recombinant adenoviruses comprising a chimeric hexon protein and/or a chimeric fiber protein, and which preferably additionally comprise a passenger gene or genes capable of being expressed in a particular cell.
  • the recombinant adenoviruses can be generated by use of a vector, specifically, a transfer vector, and preferably a viral (especially an adenoviral) or plasmid transfer vector, in accordance with the present invention.
  • a transfer vector preferably comprises a chimeric adenoviral hexon and/or fiber gene sequence as previously described.
  • a chimeric adenovirus coat protein gene sequence can simply be ligated into the vector using convenient restriction sites.
  • a wild-type adenovirus gene sequence can be mutagenized to create the chimeric coat protein sequence following its subcloning into a vector.
  • a chimeric coat protein gene sequence can be moved via standard molecular genetic techniques from a transfer vector into baculovirus or a suitable prokaryotic or eukaryotic expression vector (e.g., a viral or plasmid vector) for expression and evaluation of penton base binding, and other biochemical characteristics.
  • the present invention also provides recombinant baculoviral and prokaryotic and eukaryotic expression vectors comprising an aforementioned chimeric adenoviral coat protein gene sequence, which, along with the nucleic acid form of the adenoviral vector (i.e., an adenoviral transfer vector) are "transfer vectors" as defined herein.
  • transfer vectors By moving the chimeric gene from an adenoviral vector to baculovirus or a prokaryotic or eukaryotic expression vector, high protein expression is achievable (approximately 5-50% of the total protein being the chimeric protein) .
  • adenoviral vectors are produced using transfer vectors.
  • an adenoviral vector comprising a chimeric coat protein according to the invention can be constructed by introducing into a cell, e.g., a 293 cell, a vector comprising sequences from the adenoviral left arm, and a vector comprising sequences from the adenoviral right arm, wherein there is a region of overlap between the sequences.
  • this methodology results in recombination between the sequences, generating a vector that comprises a portion of each of the vectors, particularly the region comprising the chimeric coat protein sequences.
  • the present invention thus preferably also provides a method of constructing an adenoviral vector that has a decreased ability or inability to be recognized by a neutralizing antibody directed against wild-type adenovirus hexon protein and/or fiber protein.
  • This method comprises replacing a coat protein of the vector (i.e., a wild-type adenovirus hexon and/or fiber protein) with the corresponding chimeric adenovirus coat protein according to the invention to produce a recombinant adenoviral vector.
  • the coat protein chimera-containing particles are produced in standard cell lines, e.g., those currently used for adenoviral vectors .
  • Deletion mutants lacking the fiber gene, or possessing shortened versions of the fiber protein similarly can be employed in vector construction, e.g., H2dI802, H2dl807, H2dll021 (Falgout et al . , supra) , as can other fiber mutants.
  • the fiberless particles have been shown to be stable and capable of binding and infecting cells (Falgout et al . , supra) .
  • the present invention provides a chimeric coat protein that has a decreased ability or inability to be recognized by a neutralizing antibody directed against the corresponding wild-type coat protein, as well as vectors (including transfer vectors) comprising same.
  • the chimeric coat protein (such as a chimeric hexon and/or fiber protein) has multiple uses, e.g., as a tool for studies in vitro of capsid structure and assembly, and capsomere binding to other proteins.
  • a vector comprising a chimeric coat protein can be used in strain generation, for instance, in generation of recombinant strains of adenovirus.
  • a vector particularly an adenoviral vector, can be used in gene therapy.
  • a vector of the present invention can be used to treat any one of a number of diseases by delivering to targeted cells corrective DNA, i.e., DNA encoding a function that is either absent or impaired, or a discrete killing agent, e.g., DNA encoding a cytotoxin that, for instance, is active only intracellularly .
  • Diseases that are candidates for such treatment include, but are not limited to, cancer, e.g., melanoma, glioma or lung cancers; genetic disorders, e.g., cystic fibrosis, hemophilia or muscular dystrophy; pathogenic infections, e.g., human immunodeficiency virus, tuberculosis or hepatitis; heart disease, e.g., preventing restenosis following angioplasty or promoting angiogenesis to reperfuse necrotic tissue; and autoimmune disorders, e.g., Crohn ' s disease, colitis or rheumatoid arthritis.
  • gene therapy can be carried out in the treatment of diseases, disorders, or conditions that require repeat administration of the corrective DNA and/or the adenoviral vector, and thus for which current adenoviral-mediated approaches to gene therapy are less than optimal .
  • such a vector can be used to deliver material to a cell not as a method of gene therapy, but for diagnostic or research purposes.
  • a vector comprising a chimeric adenovirus coat protein according to the invention can be employed to deliver a gene either in vitro or in vivo, for research and/or diagnostic purposes.
  • a reporter gene or some type of marker gene can be transferred instead of transferring a so-called therapeutic gene.
  • Marker genes and reporter genes are of use, for instance, in cell differentiation and cell fate studies, as well as potentially for diagnostic purposes.
  • a standard reporter gene such as a ⁇ -galactosidase reporter gene, a gene encoding green fluorescent protein (GFP) , or a ⁇ - glucuronidase gene can be used in vivo, e.g., as a means of assay in a living host, or, for instance, as a means of targeted cell ablation (see, e.g., Minden et al .
  • transgenic animals have been- employed, for instance, for the production of recombinant polypeptides in the milk of transgenic bovine species (e.g., PCT International Application WO 93/25567) .
  • the use of an adenovirus according to the invention for gene transfer conducted for protein production in vivo further is advantageous in that such use should result in a reduced (if not absent) immune response as compared with the use of a wild-type adenovirus vector.
  • non-therapeutic reasons for gene transfer include the study of human diseases using an animal model (e.g., use of transgenic mice and other transgenic animals including p53 tumor suppressor gene knockouts for tumorigenic studies, use of a transgenic model for impaired glucose tolerance and human Alzheimer's amyloid precursor protein models for the study of glucose metabolism and for the pathogenesis of Alzheimer's disease, respectively, etc.).
  • an animal model e.g., use of transgenic mice and other transgenic animals including p53 tumor suppressor gene knockouts for tumorigenic studies, use of a transgenic model for impaired glucose tolerance and human Alzheimer's amyloid precursor protein models for the study of glucose metabolism and for the pathogenesis of Alzheimer's disease, respectively, etc.
  • an adenoviral vector comprising a chimeric adenovirus coat protein and employed as described above is advantageous in that it can be isolated and purified by conventional means. For instance, it is likely that special cell lines will not need to be made in order to propagate adenoviruses comprising the chimeric coat proteins .
  • the vectors and transfer vectors of the present invention can be employed to contact cells either in vi tro or in vivo .
  • "contacting" comprises any means by which a vector is introduced intracellularly; the method is not dependent on any particular means of introduction and is not to be so construed. Means of introduction are well known to those skilled in the art, and also are exemplified herein.
  • introduction can be effected, for instance, either in vi tro (e.g., in an ex vivo type method of gene therapy or in tissue culture studies) or in vivo by methods that include, but are not limited to, electroporation, transformation, transduction, conjugation, triparental mating, (co-) transfection, (co- ) infection, high velocity bombardment with DNA-coated microprojectiles, incubation with calcium phosphate-DNA precipitate, direct microinjection into single cells, and the like.
  • the vectors can be introduced by means of membrane fusion using cationic lipids, e.g., liposomes.
  • liposomes are commercially available (e.g., Lipofectin®, LipofectamineTM, and the like, supplied by Life Technologies, Gibco BRL, Gaithersburg, MD) .
  • liposomes having increased transfer capacity and/or reduced toxicity in vivo can be employed in the present invention.
  • Other methods also are available and are known to those skilled in the art.
  • a "host” encompasses any host into which a vector of the invention can be introduced, and thus encompasses an animal, including, but not limited to, an amphibian, bird, insect, reptile, or mammal.
  • a host is a mammal, for instance, a rodent, primate (such as chimpanzee, monkey, ape, gorilla, orangutan, or gibbon), feline, canine, ungulate (such as ruminant or swine), as well as, in particular, a human.
  • a "cell” encompasses any cell (or collection of cells) from a host into which an adenoviral vector can be introduced, e.g., preferably an epithelial cell. Any suitable organs or tissues or component cells can be targeted for vector delivery.
  • the organs/tissues/cells employed are of the circulatory system (e.g., heart, blood vessels or blood), respiratory system (e.g., nose, pharynx, larynx, trachea, bronchi, bronchioles, lungs), gastrointestinal system (e.g., mouth, pharynx, esophagus, stomach, intestines, salivary glands, pancreas, liver, gallbladder), urinary system (e.g., kidneys, ureters, urinary bladder, urethra), nervous system (e.g. brain and spinal cord, or special sense organs such as the eye) and integumentary system (e.g., skin) .
  • the cells being targeted are selected from the group consisting of heart, blood vessel, lung, liver, gallbladder, urinary bladder, and eye cells.
  • the present invention preferably also provides a method of genetically modifying a cell.
  • This method preferably comprises contacting a cell with a vector comprising a chimeric adenovirus hexon protein and/or a chimeric adenovirus fiber protein, wherein desirably the vector is an adenovirus vector.
  • the method preferably results in the production of a host cell comprising a vector according to the invention.
  • the method of the invention of genetically modifying a cell can be employed in gene therapy, or for administration for diagnosis or study.
  • the application of this method in vivo optimally comprises administering to a patient in need of gene therapy (e.g., a patient suffering from a disease, condition or disorder) a therapeutically effective amount of a recombinant adenovirus vector according to the invention.
  • This method preferably can be employed as part of an ongoing gene therapy regimen, e.g., wherein the vector (e.g., a recombinant adenovirus vector) comprising the chimeric adenovirus coat protein is administered following (e.g., after from about 1 week to about 2 months) administration of a therapeutically effective amount of a vector comprising either the corresponding wild-type coat protein or a coat protein of a different adenoviral serotype.
  • the vector comprising the chimeric adenovirus coat protein can be employed as an initial attempt at gene delivery.
  • a vector particularly an adenoviral vector of the present invention to an animal for purposes of gene therapy
  • suitable methods of administering a vector (particularly an adenoviral vector) of the present invention to an animal for purposes of gene therapy see, for example, Rosenfeld et al. (1991), supra; Jaffe et al., Clin. Res., 39 (2) , 302A (1991); Rosenfeld et al., Clin. Res., 39(2) , 311A (1991a); Berkner, supra
  • chemotherapy, vaccination, diagnosis, and/or further study are available.
  • a particular route can provide a more immediate and more effective reaction than another route.
  • local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, peritoneal, subcutaneous, intradermal, as well as topical administration.
  • Clinical trials regarding use of gene therapy vectors in vivo are ongoing. The methodology employed for such clinical trials as well as further technologies known to those skilled in the art can be used to administer the vector of the present invention for the purpose of research, diagnosis and/or gene therapy.
  • Pharmaceutically acceptable excipients also are well- known to those who are skilled in the art, and are readily available. The choice of excipient will be determined in part by the particular method used to administer the recombinant vector. Accordingly, there is a wide variety of suitable formulations for use in the context of the present invention. The following methods and excipients are merely exemplary and are in no way limiting.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules; (c) suspensions in an appropriate liquid; and (d) suitable emulsions.
  • Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • a vector of the present invention (including an adenoviral vector and a transfer vector) , alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They may also be formulated as pharmaceuticals for non-pressured preparations such as in a nebulizer or an atomizer .
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • a vector of the present invention can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • the dose administered to an animal, particularly a human, in the context of the present invention will vary with the gene of interest, the composition employed, the method of administration, the particular site and organism undergoing administration, and the reason for the administration (e.g., gene therapy, diagnosis, means of producing a protein, further study, etc) .
  • the "effective amount" of the composition is such as to produce the desired effect in a host which can be monitored using several end-points known to those skilled in the art.
  • one desired effect might comprise effective nucleic acid transfer to a host cell.
  • Such transfer can be monitored in terms of a therapeutic effect (e.g., alleviation of some symptom associated with the disease or syndrome being treated) , or by further evidence of the transferred gene or coding sequence or its expression within the host (e.g., using the polymerase chain reaction, Northern or Southern hybridizations, or transcription assays to detect the nucleic acid in host cells, or using immunoblot analysis, antibody-mediated detection, or particularized assays to detect protein or polypeptide encoded by the transferred nucleic acid, or impacted in level or function due to such transfer) .
  • a therapeutic effect e.g., alleviation of some symptom associated with the disease or syndrome being treated
  • further evidence of the transferred gene or coding sequence or its expression within the host e.g., using the polymerase chain reaction, Northern or Southern hybridizations, or transcription assays to detect the nucleic acid in host cells, or using immunoblot analysis, antibody-mediated detection, or particularized assays to detect protein or polypeptide encoded by the transferred nucle
  • the vectors of the present invention it is preferable that from about 1 to about 5,000 copies of the vector be employed per cell to be contacted, based on an approximate number of cells to be contacted in view of the given route of administration. It is even more preferable that from about 1 to about 300 plaque forming units (pfu) enter each cell.
  • this is just a general guideline which by no means precludes use of a higher or lower amount of a component, as might be warranted in a particular application, either in vi tro or in vivo .
  • the actual dose and schedule can vary depending on whether the composition is administered in combination with other pharmaceutical compositions, or depending on interindividual differences in pharmacokinetics, drug disposition, and metabolism.
  • amounts can vary in in vi tro applications depending on the particular cell type utilized or the means by which the vector is transferred.
  • One skilled in the art easily can make any necessary adjustments in accordance with the necessities of the particular situation.
  • This example describes experiments investigating adenoviral anti-vector neutralizing immunity.
  • an animal having circulating antibodies to one adenoviral vector type received intratracheal administration of another serotype adenoviral vector, and gene expression commanded by the second vector was monitored. Specifically, either an Ad4 or Ad5 wild-type vector was administered to the lungs of Sprague-Dawley rats. Ten days later, an Ad5 reporter vector was administered to the lungs of the same animals.
  • This reporter vector which is referred to herein as the "pure 5" vector, comprises an El " E3 ⁇ type 5 adenoviral vector which expresses the chloramphenicol acetyl transferase (CAT) gene driven by the cytomegalovirus early/intermediate promoter/enhancer (CMV) (i.e., AdCMVCATgD described in Kass-Eisler et al., Proc. Natl. Acad. Sci., 5, 11498-11502 (1993)).
  • CMV chloramphenicol acetyl transferase
  • CAT activity was measured in homogenized lung tissue using a CAT assay as previously described (Kass Eisler et al . (1993), supra) .
  • CAT activity was monitored at various times thereafter up to 10 days following introduction of the "pure 5" vector.
  • CAT activity was determined relative to the "pure 5" vector administered to naive animals (i.e., expression measured under this condition was considered 100%). The results of these studies are set out in Table 1, and are further reported in Mastrangeli et al . , Human Gene Therapy, 1 , 79-87 (1996) .
  • the predominant epitopes that evoke neutralizing immunity are located on the fiber and hexon, but mainly on hexon. Based on this, the effect of switching the fiber protein was investigated.
  • a vector was constructed that was identical to the "pure 5" vector except that the fiber gene was switched from a serotype 5, group C fiber to a serotype 7, group B fiber.
  • the resultant vector is referred to herein as the "5 base/7 fiber" vector.
  • the Ad5/Ad7 fiber construct was generated as shown in Figure 1. An approximately 2.7 kb (Ad5 28689-31317 bp) fragment in pAd70-100 was replaced with a Pad linker (pAd70-100dlE3.Pac) . A BamHI linker was inserted at a Muni site as indicated in Figure 2 to produce pAd70- 100dlE3.Pac.Bam. A PCR-amplified Pacl-BamHI fragment of approximately 1.1 kb containing the Ad7 fiber gene was inserted into pAd70-100dlE3. Pac .Bam to produce pAd70- 100dlE3.fiber7.
  • reporter gene assays were performed.
  • a replication-defective recombinant adenoviral reporter vector designated AdCMV- CATNeo was used in the reporter gene assay.
  • the reporter vector consists of the adenoviral origin of replication and viral packaging sequences, a combination of strong eukaryotic promoter (cytomegalovirus or CMV-1) and splicing elements, the bacterial chloramphenicol acetyl transferase (CAT) gene sequence, the mouse ⁇ maj -globin poly (A) site, the neomycin gene sequence (Neo) , and sufficient adenoviral DNA to allow for overlap recombination .
  • CMV-1 cytomegalovirus
  • CMV-1 cytomegalovirus-1
  • CAT bacterial chloramphenicol acetyl transferase
  • A mouse ⁇ maj -globin poly
  • Neo neomycin gene sequence
  • the reporter vector was used to generate AdCMV- CATNeo, AdCMV-CATNeo-dlE3 (AdCMV-CATNeo + pAd70-100dlE3) and AdCMV-CATNeo-dlE3-Fiber7 (AdCMV-CATNeo + pAd70- 1001E3. Fiber7 ) viruses.
  • Each virus was grown in large scale, i.e., a one liter suspension of human embryonic kidney 293 cells, to yield virus at a concentration of 10 12 particles/ml.
  • A549 cells were infected with an estimated 100, 300 or 1,000 particles/cell of one of the three viruses.
  • the cells were harvested and lysates were prepared as described in Kass-Eisler et al . (1993) , supra .
  • CAT assays were performed and acetylated chloramphenicol products were separated by thin layer chromatography using chloroform:methanol (95:5).
  • the results of the assays confirm that each virus was able to infect cells and express gene products at appropriate levels. Accordingly, the virus in which the native fiber was replaced with a nonnative fiber could infect cells and express genes like the parental virus .
  • This example describes the effect on neutralizing immunity of switching the fiber protein of an adenovirus from one serotype to another.
  • the "pure 5" and "5 base/7 fiber" vectors described in the preceding Example were administered to Sprague- Dawley rats which either were naive or pre-immunized against wild-type Ad5.
  • wild-type Ad5 or wild-type Ad7 (6 x 10 9 particles in phosphate buffered saline (PBS) ) was administered intraperitoneally as a primary inoculation. Seventeen days later, serum samples were taken, and about 6 x 10 9 particles in about 50 ⁇ l of PBS was injected. At about 120 hours following injection the animals were sacrificed, serum and heart tissue were harvested, and heart tissue was processed for CAT assays as previously described (Kass-Eisler et al . (1993), supra) . CAT assays also were performed on heart lysates of rat hearts infected with the "pure 5" vector or "5 base/7 fiber" vector alone.
  • This example describes the construction of adenovirus vectors wherein the neutralizing immunity-evoking epitopes have been modified.
  • this example describes vectors comprising chimeric adenoviral hexon protein, wherein the hexon neutralizing immunity-evoking epitopes are modified.
  • the results of the prior example indicate that it is possible to develop vectors for repeat administration in gene therapy from non-group C adenovirus, thus circumventing pre-existing neutralizing immunity.
  • the dominant neutralizing immunity- evoking epitopes on existing group C vectors can be modified to render the vectors less susceptible (or "stealth") to the existing neutralizing immunity.
  • adenoviral type 5-based El " E3 ⁇ CAT-expressing vectors can be constructed that have the same genetic composition as the "pure 5" and "5 base/7 fiber" vectors described above, except for possessing a gene encoding a chimeric hexon that is not recognized by pre-existing anti-type 5 neutralizing immunity.
  • the chimeric hexon gene present in the "pure 5" parental vector can be modified, in particular, II and/or 12 can be altered.
  • the hexon modifications that can be made on the "pure 5" CAT vector, or other adenoviral vector (such as any other adenoviral serotype vector), include, but are not limited to: (1) hexon with 11 deleted in its entirety; (2) hexon with 12 deleted in its entirety; (3) hexon with both 11 and 12 deleted; (4) hexon with any one or more of HVRl, HVR2, HVR3, HVR4, HVR5 , HVR6, or HVR7 , deleted; (5) -(8) hexon with a FLAG octamer epitope (i.e., Asp Tyr Lys Asp Asp Asp Asp Lys [SEQ ID NO: 50]; Hopp et al .
  • FLAG octamer epitope provides a sequence for incorporation in the chimeric hexon protein that is different from the Ad5 hexon loop sequences, and also provides a positive control using available specific anti-FLAG antibodies (Hopp et al . , supra) .
  • chimeric hexon proteins can be made in several steps.
  • a viral or plasmid vector can be constructed to contain the hexon type 5 coding sequence in a cassette that can be easily modified.
  • the hexon is read off the 1 strand of the L3 transcription unit, i.e., map units 51.6 to 59.7, comprising a region of about 2.9 kb.
  • the two other transcripts that also are encoded by L3 -- i.e., polypeptide VI and a 23 kDa protein -- do not overlap the hexon coding sequence.
  • there are no other coding sequences on the r strand that would be altered by the modification of the hexon coding sequence.
  • hexon 5 cassette comprised of an approximate 6.7 kb Sfil-Sfil fragment of the "pure 5" CAT vector.
  • Sfil cuts Ad5 into 3 fragments, the center 6.7 kb fragment (i.e., comprising about 16,282 to 22,992 base pairs, as identified by agarose gel electrophoresis) of which contains all of the L3 region plus some overlap.
  • the "hexon 5 cassette” can be subcloned into a commercially available vector having restriction sites and the like making the vector easily manipulable in terms of modification and recovery of subcloned sequences.
  • One such vector appropriate for subcloning is either the SK or KS version of the pBlueScript® phagemid (Stratagene, LaJolla, CA) .
  • the "hexon 5 cassette” can be mutagenized to generate site-specific mutations in the cloned DNA segment.
  • Several methods are available for carrying out site- specific mutagenesis.
  • the 11 and 12 deletions, insertions, or replacements (or deletions, insertions, or replacements in HVRl, HVR2, HVR3, HVR4 , HVR5, HVR6, or HVR7 regions contained therein) can be made by deleting the relevant sequences using restriction enzymes that cut uniquely within the vector inserts, or other similar means, e.g., by ligating in an end-polished, or otherwise modified, PCR product.
  • the hexon sequence contained in the hexon 5 cassette can be modified, e.g., using single-stranded mutagenesis in M13mp8 or some other convenient vector, and using appropriate oligonueleotides encompassing the flanking sequences for identification of plaques as described by Crompton et al . , supra.
  • a commercially available kit such as the ExSiteTM PCR-based site-directed mutagenesis kit and the ChameleonTM double-stranded site-directed mutagenesis kit by Stratagene can be used to introduce insertions, point mutations, or deletions into the chimeric hexon sequence without any need for subcloning into an M13, or other special vector.
  • the FLAG octapeptide sequence (Hopp et al . , supra) can be introduced into the vectors (i.e., in the presence or absence of any deletion) by inserting the relevant 24 base pair sequence (GAY TAY AAR GAY GAY GAY GAY AAR [SEQ ID NO:50], wherein Y is C or T/ ⁇ , and R is A or G) ) .
  • the replacement of Ad5 hexon loop epitopes with comparable sequences of Ad7 , Ad2, or any other adenoviral serotype, or an incorporation of these sequences in the absence of any deletion can be accomplished by using unique restriction sites, or using one of the aforementioned means of mutagenesis.
  • Ad7 neutralizing antibodies i.e., neutralizing antibodies raised in response to Ad7 innoculation of a naive animal
  • both hypervariable loops 1 and 2 can be deleted from a serotype 5 or another serotype adenoviral vector.
  • Adenoviral vectors and there genomes comprising these deletions are useful as a starting point to create other adenoviral vectors having loop replacements, as a tool for studying hexon structure-function relationships, and under some circumstances as a gene transfer vector with limited vulnerability to the adaptive immune system.
  • This example describes the method of replacing the hexon protein of one serotype adenoviral vector with the hexon protein of another serotype adenoviral vector to generate a recombinant adenovirus .
  • the hexon protein of an Ad5 vector was replaced with the hexon protein of an Ad2 vector.
  • This example also describes the method of incorporating the chimeric hexon proteins of the preceding Example into a vector to make a recombinant adenovirus.
  • Ad5 hexon gene open reading frame ORF
  • Ad2 hexon gene ORF Ad2 hexon gene open reading frame
  • Adenoviral vectors comprising modified or chimeric hexon proteins can be constructed by homologous recombination using standard techniques and human embryonic kidney 293 cells (see, e.g., Rosenfeld et al . (1991), supra; Rosenfeld et al . (1992), supra) .
  • map units 0 to 57.3 of dlAd5NCAT can be isolated by Bsu36l digestion
  • map units 58.4 to 100 of dlAd5NCAT can be isolated by Drdl digestion.
  • a neutralizing antibody directed against the parental vector can be employed to facilitate the generation of hexon replacement constructs.
  • anti-Ad5 neutralizing polyclonal or monoclonal antibodies directed against the loops 1 and 2 of Ad5 hexon
  • the presence of the Ad5 neutralizing antibodies substantially blocks the propagation of the undesired wildtype Ad5 vector (s), while the chimeric vector is unaffected.
  • the recombinant vectors comprising a chimeric hexon ORF can be generated by homologous recombination using a plasmid that carries a marker gene, such as Green Fluorescent Protein (GFP) , adjacent to the chimeric or novel hexon ORF (e.g., between the fiber and hexon genes) .
  • a marker gene such as Green Fluorescent Protein (GFP)
  • GFP Green Fluorescent Protein
  • genomes that could harbor the chimeric hexon gene should also harbor the marker gene.
  • the marker gene would then be expressed as a late protein, so that cells that potentially comprise the desired adenoviral genome can be easily identified.
  • vectors can be constructed that have the aforementioned hexon modifications, and which have further modifications, for instance, in the adenoviral fiber coding sequences. This can be accomplished by making the hexon modifications described above, and using different parental plasmids for homologous recombination, such as parental plasmids comprising mutations in fiber coding sequences.
  • the "5 base/7 fiber" vector can be employed as a starting vector for vector construction.
  • All of the viral vectors prepared according to this example can be plaque-purified, amplified, and further purified using standard methods (Rosenfeld et al . (1991), supra; Rosenfeld et al . (1992), supra) .
  • This example describes a characterization of the activity in vi tro and in vivo of the vectors described in the preceding Examples.
  • each of the viruses prepared as described in the preceding Examples can be evaluated in vi tro and in vivo using standard methods as previously described (e.g., Kass-Eisler et al . , supra) , and as set forth herein.
  • the various vectors along with control vectors e.g., the "pure 5" and "5 base/7 fiber” vectors, and the Ad5 wild-type vector
  • the various vectors along with control vectors e.g., the "pure 5" and "5 base/7 fiber” vectors, and the Ad5 wild-type vector
  • the various vectors e.g., the "pure 5" and "5 base/7 fiber” vectors, and the Ad5 wild-type vector
  • the cells are then evaluated for
  • the in vivo studies can be carried out in Sprague- Dawley rats.
  • the Sprague-Dawley rat as opposed to the mouse or cotton rat is preferred for these experiments since the rat is non-permissive, and the wild-type adenovirus cannot replicate in this host.
  • immunizations can be carried out using wild-type viruses (e.g., wild-type Ad5 or Ad7 ) , the "pure 5" CAT vector, and the "5 base/7 fiber" CAT vector by intravenous administration (e.g., Kass-Eisler et al . , supra) .
  • the vector of interest can be administered intravenously or directly into the airways of the host.
  • intravenous administration allows an assessment of the "worst case scenario" (i.e., wherein the vector is in immediate contact with the circulating humoral immune system, and thus the strongest immune response is to be expected)
  • introduction in the airways of the host allows an evaluation of a compartmentalized and mucosal humoral immune response.
  • CAT activity can be quantified as previously described in all the relevant organs, e.g., liver, heart, and lung for intravenous administration, and lung only for respiratory administration. Appropriate standards can-be used to compensate for variations in organ expression of CAT activity (see e.g., Kass-Eisler et al . , Gene Therapy, 2_ 395-402 (1994)) . The in vitro and in vivo results can be compared and assessed using standard statistical methods .
  • NAME CORNELL RESEARCH FOUNDATION, INC.
  • NAME GENVEC, INC.
  • NAME CRYSTAL, RONALD G.
  • NAME GALL, JASON
  • NAME KOVESDI, IMRE
  • NAME WICKHAM, THOMAS J.
  • MOLECULE TYPE DNA (genomic)
  • ATA GGA TCA GAC AAT GCA GAA ACA CAA GCT AAA CCT GTA TAC GCA GAT 624 lie Gly Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp 195 200 205
  • GGC AAT GCT ACT AAA CCA AAA GTG GTT TTG TAC AGT GAA GAT GTA AAT 912 Gly Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn 290 295 300
  • CAG GTG CCC CAA AAG TTT TTT GCC ATT AAA
  • MOLECULE TYPE DNA (genomic)
  • GCT TCC ACG TAC TTT GAC ATC CGC GGC GTG CTG GAC AGG GGC CCT ACT 336 Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
  • GCC CCA AAT CCT TGC GAA TGG GAT GAA GCT GCT ACT GCT CTT GAA ATA 432 Ala Pro Asn Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He 130 135 140 AAC CTA GAA GAA GAG GAC GAT GAC AAC GAA GAC GAA GTA GAC GAG CAA 480 Asn Leu Glu Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin 145 150 155 160
  • AAA AAT GAA ATA AGA GTT GGA AAT AAT TTT GCC ATG GAA ATC AAT CTA 1392 Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 450 455 460
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO: 39:
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • GGC AAT GCT ACT AAA CCA AAA GTG GTT TTG TAC AGT GAA GAT GTA AAT 912 Gly Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn 290 295 300
  • CAG GTG CCC CAA AAG TTT TTT GCC ATT AAA

Abstract

The present invention provides a chimeric adenoviral coat protein (particularly a chimeric adenovirus hexon protein). The chimeric adenovirus coat protein has a decreased ability or inability to be recognized by a neutralizing antibody directed against the corresponding wild-type adenovirus coat protein.

Description

CHIMERIC ADENOVIRAL COAT PROTEIN AND METHODS OF USING SAME
TECHNICAL FIELD OF THE INVENTION
The present invention relates to a chimeric adenoviral coat protein and a recombinant adenovirus comprising same. In particular, the invention provides a chimeric adenoviral hexon protein and a recombinant adenovirus comprising the chimeric adenoviral hexon protein. Such a recombinant adenovirus can be employed inter alia in gene therapy.
BACKGROUND OF THE INVENTION
In vivo gene therapy is a strategy in which nucleic acid, usually in the form of DNA, is administered to modify the genetic repertoire of target cells for therapeutic purposes. This can be accomplished efficiently using a recombinant adenoviral vector encoding a so-called "therapeutic gene". A therapeutic gene is generally considered a gene that corrects or compensates for an underlying protein deficit or, alternately, a gene that is capable of down-regulating a particular gene, or counteracting the negative effects of its encoded product, in a given disease state or syndrome. Recombinant adenoviral vectors have been used to transfer one or more recombinant genes to diseased cells or tissues in need of treatment. As reviewed by Crystal, Science, 270, 404-410 (1995), such vectors are preferred over other vectors commonly employed for gene therapy (e.g., retroviral vectors) since adenoviral vectors can be produced in high titers (i.e., up to 1013 viral particles/ml), and they efficiently transfer genes to nonreplicating, as well as replicating, cells. Moreover, adenoviral vectors are additionally preferred based on their normal tropism for the respiratory epithelium in cases where the targeted tissue for somatic gene therapy is the lung, as well as for other reasons (see, e.g., Straus, Tn Adenoviruses, Plenan Press, New York, NY, 451-496 (1984)); Horwitz et al . , I_n Virology, 2nd Ed., Fields et al . , eds., Raven Press, New York, NY, 1679-1721 (1990); Berkner, BioTechniques, _6, 616 (1988); Chanock et al . , JAMA, 195, 151 (1966); Haj-Ahmad et al . , J. Virol., 57_, 267 (1986); and Ballay et al . , EMBO, -4, 3861 (1985)).
There are 49 human adenoviral serotypes, categorized into 6 subgenera (A through F) based on nucleic acid comparisons, fiber protein characteristics, and biological properties (Crawford-Miksza et al . , J. Virol., 70, 1836- 1844 (1996)). The group C viruses (e.g., serotypes 2 and 5, or Ad2 and Ad5) are well characterized. It is these serotypes that currently are employed for gene transfer studies, including human gene therapy trials (see, e.g., Rosenfeld et al . , Science, 252, 431-434 (1991); Rosenfeld et al., Cell, _L, 143-155 (1992); Zabner, Cell, 7_5, 207- 216 (1993); Crystal et al . , Nat. Gen., 8_, 42-51 (1994); Yei et al., Gene Therapy, 1, 192-200 (1994); Chen et al . , Proc. Natl. Acad. Sci . , 91, 3054-3057 (1994); Yang et al., Nat. Gen., 1_, 362-369 (1994); Zabner et al . , Nat. Gen., 6 , 75-83 (1994)). Other groups and serotypes include, but are not limited to: group A (e.g., serotypes 12 and 31), group B (e.g., serotypes 3 and 7), group D (e.g., serotypes 8 and 30), group E (e.g., serotype 4) and group F (e.g., serotypes 40 and 41) (Horwitz et al . , supra) .
In terms of general structure, all adenoviruses examined to date are nonenveloped, regular icosahedrons of about 65 to 80 nanometers in diameter. Adenoviruses are comprised of linear, double-stranded DNA that is complexed with core proteins and surrounded by the adenoviral capsid. The capsid is comprised of 252 capsomeres, of which 240 are hexons and 12 are pentons . The hexon capsomere provides structure and form to the capsid (Pettersson, in The Adenoviruses, pp. 205-270, Ginsberg, ed., (Plenum Press, New York, NY, 1984)), and is a homotrimer of the hexon protein (Roberts et al . , Science, 232, 1148-1151 (1986)). The penton comprises a penton base, which is bound to other hexon capsomeres, and a fiber, which is noncovalently bound to, and projects from, the penton base. The penton fiber protein comprises three identical polypeptides (i.e., polypeptide IV). The Ad2 penton base protein comprises five identical polypeptides (i.e., polypeptide III) of 571 amino acids each (Boudin et al., Virology, 92, 125-138 (1979)).
The adenoviruses provide an elegant and efficient means of transferring therapeutic genes into cells. However, one problem encountered with the use of adenoviral vectors for gene transfer in vivo is the generation of antibodies to antigenic epitopes on adenoviral capsid proteins. If sufficient in titer, the antibodies can limit the ability of the vector to be used more than once as an effective gene transfer vehicle. For instance, animal studies demonstrate that intravenous or local administration (e.g., to the lung, heart or peritoneum) of an adenoviral type 2 or 5 gene transfer vector can result in the production of antibodies directed against the vector which prevent expression from the same serotype vector administered 1 to 2 weeks later (see, e.g., Yei et al., supra; Zabner (1994), supra; Setoguchi et al . , Am. J. Respir. Cell. Mol. Biol., 10, 369-377 (1994); Kass-Eisler et al . , Gene Therapy, 1, 395- 402 (1994); Kass-Eisler et al . , Gene Therapy 3, 154-162 (1996)). This is a drawback in adenoviral-mediated gene therapy, since many uses of an adenoviral vector (e.g., for prolonged gene therapy) require repeat administration inasmuch as the vector does not stably integrate into the host cell genome. The mechanism by which antibodies directed against an adenovirus are able to prevent or reduce expression of an adenoviral-encoded gene is unclear. However, the phenomenon is loosely referred to as "neutralization", and the responsible antibodies are termed "neutralizing antibodies."
There are three capsid structures against which neutralizing antibodies potentially can be elicited: fiber, penton, and hexon (Pettersson, supra) . The hexon protein, and to a lesser extent the fiber protein, comprise the main antigenic determinants of the virus, and also determine the serotype specificity of the virus (Watson et al . , J. Gen. Virol., 69, 525-535 (1988); Wolfort et al., J. Virol., 62:, 2321-2328 (1988); Wolfort et al., J. Virol., 5_6, 896-903 (1985); Crawford-Miksza et al . , supra) . Researchers have examined and compared the structure of these coat proteins of different adenoviral serotypes in an effort to define the regions of the proteins against which neutralizing antibodies are elicited.
The Ad2 hexon trimer is comprised of a pseudohexagonal base and a triangular top formed of three towers (Roberts et al . , supra; Athappilly et al . , J. Mol. Biol. , 242, 430-455 (1994)). The base pedestal consists of two tightly packed eight-stranded antiparallel beta barrels stabilized by an internal loop. The predominant regions in hexon protein against which neutralizing antibodies are directed appear to be in loops 1 and 2 (i.e., LI or 11, and LII or 12 , respectively) in one of the three towers. For instance, Kinloch et al . (J. Biol. Chem. , 258, 6431-6436 (1984)) compared adenoviral hexon sequences and theorized that the serotype-specific antigenic determinants on hexon are located in amino acid residues 120 to 470 encompassing the 11 and 12 loops since type-specific sequence differences are mainly concentrated in this region. Toogood et al . (J. Gen. Virol. , 73, 1429-1435 (1992)) used peptides from this region to generate specific anti-loop antisera and confirmed that antibodies against residues 281-292 of 11 and against residues 441-455 of 12 were sufficient to neutralize infection. Also, Crompton et al . (J. Gen. Virol. , 7_5, 133-139 (1994)) modified these loops to accept neutralizing epitopes from polio virus, and demonstrated that infection with the resultant adenoviral vector generated neutralizing immunity against polio virus. More recently it was demonstrated that the hexon protein is composed of seven discrete hypervariable regions in loops and 1 and 2 (HVRl to HVR7 ) which vary in length and sequence between adenoviral serotypes (Crawford-Miksza et al., supra) .
Less is known regarding the regions of the fiber protein against which neutralizing antibodies potentially can be directed. However, much data is available on the structure of the fiber protein. The trimeric fiber protein consists of a tail, a shaft, and a knob (Devaux et al., J. Molec. Biol., 215, 567-588 (1990)). The fiber shaft region is comprised of repeating 15 amino acid motifs, which are believed to form two alternating beta strands and beta bends (Green et al . , EMBO J . , 2_, 1357- 1365 (1983)). The overall length of the fiber shaft region and the number of 15 amino acid repeats differ between adenoviral serotypes. The receptor binding domain of the fiber protein and sequences necessary for fiber trimerization are localized in the knob region encoded by roughly the last 200 amino acids of the protein (Henry et al., J. Virol., 68 (8) , 5239-5246 (1994)); Xia et al., Structure, 2(12), 1259-1270 (1994)). Furthermore, all adenovirus serotypes appear to possess a type of specific moiety located in the knob region (Toogood et al . , supra . )
Given the existence of these potential epitopes in hexon protein and fiber protein, it is understandable that, in some cases, difficulties have been encountered using adenovirus as a vector for gene therapy. Accordingly, recombinant adenoviral vectors capable of escaping such neutralizing antibodies (in the event they are preexisting and hamper gene expression commanded by adenovirus in an initial dose) , and which would allow repeat doses of adenoviral vectors to be administered, would significantly advance current gene therapy methodology.
Thus, the present invention seeks to overcome at least some of the aforesaid problems of recombinant adenoviral gene therapy. In particular, it is an object of the present invention to provide a recombinant adenovirus comprising a chimeric coat protein that has a decreased ability or inability to be recognized by antibodies (i.e., neutralizing antibodies) directed against the corresponding wild-type adenovirus coat protein. These and other objects and advantages of the present invention, as well as additional inventive features, will be apparent from the description of the invention provided herein.
BRIEF SUMMARY OF THE INVENTION
The present invention provides a chimeric adenovirus coat protein (particularly a chimeric adenovirus hexon protein) comprising a nonnative amino acid sequence. The chimeric adenovirus coat protein is not recognized by, or has a decreased ability to be recognized by, a neutralizing antibody directed against the corresponding wild-type (i.e., native) coat protein. The chimeric adenovirus coat protein enables a vector (such as an adenovirus) comprising the corresponding protein to be administered repetitively, or to be administered following administration of an adenovirus vector comprising the corresponding wild-type coat protein. It also enables a vector (such as an adenovirus) comprising the chimeric protein to be administered and effect gene expression in the case where there are preexisting neutralizing antibodies directed against the wild-type adenovirus coat protein. The present invention also provides a vector, particularly an adenoviral vector, that comprises a chimeric adenovirus coat protein such as chimeric adenovirus hexon protein (and which optionally further comprises a chimeric adenovirus fiber and/or penton base protein) , and methods of constructing and using such a vector .
BRIEF DESCRIPTION OF THE FIGURES Figure 1 is a diagram of the method employed to construct the vector pAd70-100dlE3. fiber7.
Figure 2 is a partial restriction map of the vector pGBS.59-100(HSF:RGD) .
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides, among other things, a chimeric adenovirus coat protein. The chimeric adenovirus coat protein comprises a nonnative amino acid sequence, such that the chimeric adenovirus coat protein (or a vector comprising the chimeric adenovirus coat protein) has a decreased ability or inability to be recognized by antibodies (e.g., neutralizing antibodies) directed against the corresponding wild-type adenovirus coat protein .
Chimeric Adenovirus Coat Protein
A "coat protein" according to the invention is either an adenoviral penton base protein, an adenoviral hexon protein, or an adenoviral fiber protein. Preferably a coat protein is a adenoviral hexon protein or an adenoviral fiber protein. Any one of the serotypes of human or nonhuman adenovirus can be used as the source of the coat protein, or its gene or coding sequence. Optimally, however, the adenovirus coat protein is that of a Group B or C adenovirus and, preferably, is that of Adl, Ad2, Ad3, Ad5, Ad6, Ad7 , Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, or Ad48.
The chimeric adenovirus coat protein (or a vector, such as adenoviral vector, comprising the chimeric adenovirus coat protein) has a decreased ability or an inability to be recognized by an antibody (e.g., a neutralizing antibody) directed against the corresponding wild-type adenovirus coat protein. A "neutralizing antibody" is an antibody that either is purified from or is present in serum. As used herein, an antibody can be a single antibody or a plurality of antibodies. An antibody is "neutralizing" if it inhibits infectivity of (i.e., cell entry) or gene expression commanded by an adenovirus comprising wild-type coat protein, or if it exerts a substantial deleterious effect on infectivity of or gene expression commanded by an adenovirus comprising wild-type coat protein, as compared, for instance, to any effect on any other adenoviral property.
An ability or inability of a chimeric coat protein to "be recognized by" (i.e., interact with) a neutralizing antibody directed against the wild-type adenovirus coat protein can be assessed by a variety of means known to those skilled in the art. For instance, the removal of one or more epitopes for a neutralizing antibody present in a wild-type adenovirus coat protein to generate a chimeric adenovirus coat protein will result in a decreased ability or inability of the chimeric coat protein to be recognized by the neutralizing antibody. Also, such a decreased ability or inability to interact with a neutralizing antibody directed against wild-type coat protein can be demonstrated by means of a neutralization test (see, e.g., Toogood et al . , supra; Crawford-Miksza et al . , supra; Mastrangeli et al., Human Gene Therapy, 1_, 79-87 (1996) ) , or as further described herein .
Generally, an "inability" of a chimeric adenovirus coat protein (or a vector comprising a chimeric adenovirus coat protein) to be recognized by a neutralizing antibody directed against wild-type adenovirus coat protein means that such an antibody does not interact with the chimeric coat protein, and/or exhibits no substantial deleterious effect on infectivity of or gene expression commanded by an adenovirus comprising wild-type coat protein, as compared, for instance, to any effect on any other adenoviral property.
A "decreased ability" to be recognized by neutralizing antibody directed against wild-type adenovirus coat protein refers to any decrease in the ability of the chimeric adenovirus coat protein (or a vector comprising the chimeric coat protein) to be recognized by an antibody directed against the corresponding wild-type adenovirus coat protein as compared to the wild-type adenovirus coat protein. When such ability/inability is assessed by means of a neutralization test in particular, preferably a "decreased ability" to be recognized by a neutralizing antibody directed against wild-type adenovirus coat protein is exhibited by from about a 10% to about a 99% increase in the ability of a recombinant adenovirus comprising the chimeric coat protein to cause a visible cytopathic effect (c.p.e.) in cells such as A549 cells or COS-1 cells (or other such cells appropriate for a neutralization assay) in the presence of the neutralizing antibody as compared to an adenovirus comprising the wild-type coat protein against which the neutralizing antibody is directed. Furthermore, a decreased ability or inability of an adenovirus chimeric coat protein (or a vector comprising a chimeric adenovirus coat protein) to interact with a neutralizing antibody can be shown by a reduction of inhibition (from about 10% to about 99%) or no inhibition at all of cell infectivity by a recombinant vector (such as an adenoviral vector) containing the chimeric coat protein as compared to a recombinant vector containing the wild-type protein. Also, a decreased ability or inability of an adenovirus chimeric coat protein (or a vector comprising a chimeric adenovirus coat protein) to interact with a neutralizing antibody can be shown by a reduction of inhibition (from about 10% to about 99%) or no inhibition at all of gene expression commanded by a recombinant vector (such as an adenoviral vector) containing the chimeric coat protein as compared to a recombinant vector containing the wild-type coat protein. These tests can be carried out when the recombinant adenovirus containing the chimeric coat protein is administered following the administration of an adenovirus containing the wild-type coat protein, or when the recombinant adenovirus is administered to a host that has never before encountered or internalized adenovirus (i.e., a "naive" host) . These methods are described, for instance, in the Examples which follow as well as in Mastrangeli et al . , supra . Other means such as are known to those skilled in the art also can be employed.
The coat protein is "chimeric" in that it comprises a sequence of amino acid residues that is not typically found in the protein as isolated from, or identified in, wild-type adenovirus, which comprises the so-called native coat protein, or "wild-type coat protein" . The chimeric coat protein thus comprises (or has) a "nonnative amino acid sequence". By "nonnative amino acid sequence" is meant any amino acid sequence (i.e., either component residues or order thereof) that is not found in the native coat protein of a given serotype of adenovirus, and which preferably is introduced into the coat protein at the level of gene expression (i.e., by production of a nucleic acid sequence that encodes the nonnative amino acid sequence) . Generally, the nonnative amino acid sequence can be obtained by deleting a portion of the amino acid sequence, deleting a portion of the amino acid sequence and replacing the deleted amino sequence with a so-called "spacer region", or introducing the spacer region into an unmodified coat protein. Preferably such manipulations result in a chimeric adenovirus coat protein according to the invention that is capable of carrying out the functions of the corresponding wild-type adenovirus coat protein (or, at least that when incorporated into an adenovirus, will allow appropriate virion formation and will not preclude adenoviral-mediated cell entry) , and, optimally, that is not impeded in its proper folding. Also, it is desirable that the manipulations do not result in the creation of new epitopes for differing antibodies, unless, of course, such epitopes do not interfere with use of an adenovirus containing the chimeric coat protein as a gene transfer vehicle in vivo .
In particular, a nonnative amino acid sequence according to the invention preferably comprises a deletion of a region of a wild-type adenovirus coat protein, particularly an adenovirus hexon or fiber protein. Optimally the resultant nonnative amino acid sequence is such that one or more of the existing epitopes for neutralizing antibodies directed against the corresponding wild-type adenovirus coat protein have been rendered non- immunogenic. Desirably, the region deleted comprises from about 1 to about 750 amino acids, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted comprises a smaller region less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids. The chimeric coat protein also desirably comprises a plurality of such deletions. Thus, according to the invention, the chimeric adenovirus coat protein comprises modification of one or more amino acids, and such modification is made in one or more regions .
In a preferred embodiment of the present invention, a nonnative amino acid sequence comprises a deletion of one or more regions of a wild-type adenovirus hexon protein, wherein preferably the hexon protein is the Ad2 hexon protein [SEQ ID NO: 2] (which is encoded by the sequence of SEQ ID NO:l; GenBank® Data Bank Accession Number U20821), or the Ad5 hexon protein [SEQ ID NO: 3] (GenBank® Data Bank Accession Number M73260, which is encoded by the sequence of SEQ ID NO:4), or the Ad7 hexon protein (GenBank® Data Bank Accession Number x76551) . Alternately, preferably the hexon protein is the protein sequence reported by Crawford-Miksza et al . (Ad2 hexon [SEQ ID NO:52], Ad5 hexon SEQ ID NO:54]) . In particular, the sequences of Crawford-Miksza et al . differ over those reported in the GenBank® Data Bank in that the amino acid residue reported as the first in the Crawford-Miksza et al . sequences is not Met, and the Ad5 hexon sequence is reported as terminating with "Gin His" instead of with "Thr Thr" . As employed herein, the numbering of adenovirus hexon amino acid residues corresponds to that in Crawford-Miksza et al.
Desirably the region (s) of the deletion comprises an internal hexon protein sequence ("internal" meaning not at or near the C- or N-terminus of the protein; "near" referring to a distance of 500 amino acids or less ) , preferably a hypervariable region, e.g., as reported in Crawford-Miksza et al . In particular, optimally, the internal region of the wild-type hexon protein that is deleted to generate the chimeric hexon protein comprises the entirety of 11 loop, preferably from about residue 131 to about residue 331 of the Ad2 hexon protein [SEQ ID NO: 6] (which is encoded by the sequence of SEQ ID NO: 5), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad5 [SEQ ID NO: 8] (which is encoded by the sequence of SEQ ID NO:7), Ad6, Ad7 , Ad8, Adl2, Adlδ, Ad40, Ad41, Ad48, BAV3, or MAV1, especially as reported in Crawford- Miksza et al . , supra .
Alternately, preferably the internal region of the wild-type hexon protein that is deleted to produce the chimeric hexon protein comprises one or more regions (e.g., smaller regions) of the 11 loop. Optimally the region deleted comprises a hypervariable region. Desirably the one or more regions of the II loop deleted are regions (i.e., hypervariable regions) selected from this group consisting of the HVRl region, the HVR2 region, the HVR3 region, the HVR4 region, the HVR5 region, and the HVR6 region. Moreover, preferably the region of the wild- type protein that is deleted (or otherwise manipulated as described herein) occurs on the external surface of the hexon protein. Thus, HVR2, HVR3, HVR4 , and HVR5 — each of which are externally located regions of the hexon protein -- are particularly preferred for deletion or modification .
The "HVRl region" preferably comprises from about amino acid 137 to about amino acid 188 of the Ad2 hexon protein [SEQ ID NO: 10] (which is encoded by the sequence of SEQ ID NO: 9), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 12] (which is encoded by the sequence of SEQ ID NO:ll), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
The "HVR2 region" preferably comprises from about amino acid 194 to about amino acid 204 of the Ad2 hexon protein [SEQ ID NO: 14] (which is encoded by the sequence of SEQ ID NO: 13), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 16] (which is encoded by the sequence of SEQ ID NO:15), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
The "HVR3 region" preferably comprises from about amino acid 222 to about amino acid 229 of the Ad2 hexon protein [SEQ ID NO: 18] (which is encoded by the sequence of SEQ ID NO:17), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO:20] (which is encoded by the sequence of SEQ ID NO:19), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adlδ, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
The "HVR4 region" preferably comprises from about amino acid 258 to about amino acid 271 of the Ad2 hexon protein [SEQ ID NO: 22] (which is encoded by the sequence of SEQ ID NO:21), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 24] (which is encoded by the sequence of SEQ ID NO:23), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
The "HVR5 region" preferably comprises from about amino acid 278 to about amino acid 294 of the Ad2 hexon protein [SEQ ID NO:26] (which is encoded by the sequence of SEQ ID NO:25), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO:28] (which is encoded by the sequence of SEQ ID NO:27), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra . In particular, preferably the deleted region comprises from about amino acid 297 to about amino acid 304 just outside of the HVR5 region of the Ad2 hexon protein [SEQ ID NO: 30] (which is encoded by the sequence of SEQ ID NO:29), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 32] (which is encoded by the sequence of SEQ ID NO:31), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
The "HVR6 region" preferably comprises from about amino acid 316 to about amino acid 327 of the Ad2 hexon protein [SEQ ID NO: 34] (which is encoded by the sequence of SEQ ID NO:33), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 36] (which is encoded by the sequence of SEQ ID NO:35), Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra .
In another preferred embodiment of the invention, the internal region of the wild-type hexon protein that is deleted to generate the chimeric hexon protein comprises the entirety of the 12 loop, preferably from about residue 423 to about residue 477 of the Ad2 hexon protein [SEQ ID NO:38] (which is encoded by the sequence of SEQ ID NO:37), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 40] (which is encoded by the sequence of SEQ ID NO:39), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra . Alternately, preferably the internal region of the wild-type hexon protein that is deleted to produce the chimeric hexon protein comprises one or more smaller regions (e.g., hypervariable regions) of the 12 loop. In particular, preferably the smaller region of the 12 loop comprises the HVR7 region.
The "HVR7 region" preferably comprises from about amino acid 433 to about amino acid 465 of the Ad2 hexon protein [SEQ ID NO: 42] (which is encoded by the sequence of SEQ ID NO:41), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 44] (which is encoded by the sequence of SEQ ID NO:43), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra . In particular, preferably the deleted region comprises from about amino acid 460 to about amino acid 466 of the HVR7 region (i.e., extending one base pair outside of this region) of the Ad2 hexon protein [SEQ ID NO:46] (which is encoded by the sequence of SEQ ID NO:45), or the corresponding region from another adenoviral serotype, e.g., particularly the corresponding region from Adl, Ad3, Ad5 [SEQ ID NO: 48] (which is encoded by the sequence of SEQ ID NO:47), Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al., supra .
Along the same lines, the chimeric adenovirus hexon protein desirably comprises deletions in one or both of the aforementioned regions, i.e., the hexon protein comprises deletions in one or both of the 11 and 12 loops, which deletions can constitute the entirety of the loop(s), or can comprise deletions of one or more smaller regions (e.g., hypervariable regions) in one or both of the hexon loops. In particular, de.sirably the deleted region (s) are selected from the group consisting of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO: 46, and SEQ ID NO: 48, and equivalents and conservative variations of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48.
An "equivalent" is a naturally occurring variation of an amino acid or nucleic acid sequence, e.g., as are observed among different strains of adenovirus. A conservative variation is a variation of an amino acid sequence that results in one or more conservative amino acid substitution (s) . A "conservative amino acid substitution" is an amino acid substituted by an alternative amino acid of similar charge density, hydrophilicity/hydrophobicity, size, and/or configuration
(e.g., basic, Arg and Lys; aliphatic Ala, Cys, Gly, lie, Leu, Met and Val; aromatic, Phe, Tyr, Trp, and His; hydrophilic, Glu, Gin, Asn, and Asp; hydroxyl, Ser and Thr) .
In another preferred embodiment, the nonnative amino acid sequence of the chimeric adenoviral coat protein
(i.e., particularly a chimeric adenoviral fiber or hexon protein) comprises a deletion of one or more region (s) of the wild-type adenovirus coat protein (particularly the 11 and/or 12 loops, and, most particularly, the HVRl, HVR2, HVR3, HVR4, HVR5, HVR6, and/or HVR7 regions of the wild- type adenovirus hexon protein) as previously described, and further comprises a replacement of the region (s) with a spacer region preferably of from 1 to about 750 amino acids, especially of from about 1 to about 500 amino acids, and particularly of from about 1 to about 300 amino acids. It also is desirable that the region deleted and replaced comprises a smaller region less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids. The chimeric coat protein also desirably comprises a plurality of such replacements. Thus, according to the invention, the chimeric adenovirus coat protein comprises modification of one or more amino acids, and such modification is made in one or more regions which can be a smaller region. A spacer region of the aforementioned size also preferably simply can be inserted into one of the aforementioned regions (particularly into the 11 and/or 12 loop, or one or more of the aforementioned HVRl, HVR2, HVR3, HVR4 , HVR5, HVR6, and HVR7 regions of the adenovirus hexon protein) in the absence of any deletion to render the resultant chimeric protein nonimmunogenic by, for instance, destroying the ability of a neutralizing antibody to interact with that particular site (e.g., by changing the spatial juxtaposition of critical amino acids with which the antibody interacts) .
Optimally the spacer region comprises a nonconservative variation of the amino acid sequence of wild-type adenovirus coat protein (particularly wild-type adenovirus hexon protein) that comprises an epitope for a neutralizing antibody, and which may or may not be deleted upon the insertion of the spacer region. A "nonconservative variation" is a variation of this amino acid sequence that does not result in the creation or recreation in the chimeric adenovirus coat protein of the epitope for a neutralizing antibody directed against the wild-type adenovirus coat protein, and, in particular, is a variation of the spacer region that results in one or more nonconservative amino acid insertion (s) or substitution (s) in this region. A "nonconservative amino acid substitution" is an amino acid substituted by an alternative amino acid of differing charge density, hydrophilicity/hydrophobicity, size, and/or configuration (e.g., a change of a basic amino acid for an acidic amino acid, a hydrophilic amino acid for a hydrophobic amino acid, and the like) .
Desirably the spacer region does not interfere with the functionality of the chimeric adenovirus coat protein, particularly the chimeric adenovirus hexon or fiber protein, e.g., the ability of hexon protein to bind penton base protein or other hexon capsomeres, or the ability of penton fiber to bind penton base and/or to a cell surface receptor. Such functionality can be assessed by virus viability. Similarly, the absence of the creation or recreation of the epitope (s) for a neutralizing antibody directed against the wild-type coat (e.g., hexon and/or fiber) protein can be confirmed using techniques as described in the Examples which follow (e.g., by ensuring the antibody, which may be in a carrier fluid such as . serum or other liquid, binds the wild-type adenovirus coat protein, but not the chimeric adenovirus coat protein) .
Preferably the spacer region incorporated into the adenovirus coat protein (i.e., either as an insertion into the wild-type coat protein, or to replace one or more deleted region (s) of the wild-type adenovirus coat protein) comprise a series of polar and/or charged amino acids (e.g., Lys, Arg, His, Glu, Asp, and the like), or amino acids with intermediate polarity (e.g., Gin, Asn, Thr, Ser, Met, and the like) . In particular, desirably the spacer region comprises the sequence of SEQ ID NO: 50 (which is encoded by the sequence of SEQ ID NO:49), and equivalents and conservative variations of SEQ ID NO: 50. Alternately, the spacer region can comprise any other sequence like the FLAG octapeptide sequence of SEQ ID NO: 50 that will not interfere with the functionality of the resultant chimeric protein.
In still yet another preferred embodiment, a region of a wild-type adenovirus coat protein (particularly an adenovirus hexon and/or fiber protein) is deleted and replaced with a spacer region comprising the corresponding coat protein region of another adenoviral serotype. Preferably in this embodiment the spacer region is of a different adenoviral group. For instance, preferably a region of an Ad2 coat protein can be replaced with the corresponding region of an Ad5 or Ad7 coat protein (or any other serotype of adenovirus as described above) , and vice versa. It also is preferable that such a spacer region comprising the coat protein region of another adenoviral serotype is simply inserted into the corresponding coat protein region of the chimeric coat protein. In this case, the likelihood of obtaining a chimeric hexon protein that is functional can be increased by making sure that the size of the hypervariable domain resulting from such insertion approximates the size of a known hypervariable domain. For instance, the HVRl region of Ad40 is about 30 amino acids smaller than the HVRl region of Ad2 (as well as other adenoviruses such as Ad5, Ad8, etc.) . Thus, preferably a spacer region of about 30 amino acids can be incorporated into the Ad40 HVRl region to produce a chimeric adenovirus hexon protein. In particular, desirably the region of Ad2 (or other adenovirus) that is not present in Ad40 (i.e., approximately amino acid residues 138 to 174), or a portion thereof, is introduced into Ad40 to produce the chimeric adenoviral hexon protein .
According to the invention, desirably the nonnative amino acid sequence of a chimeric coat protein comprises a plurality of such replacements or insertions. When the coat protein is incorporated into an adenoviral vector, preferably the entire coat protein of one adenoviral serotype can be substituted with the entire coat protein of another adenoviral serotype, as described further herein .
The region or regions of wild-type adenovirus hexon protein that are deleted and replaced by the spacer region, or into which the spacer region is inserted, can be any suitable region (s) and desirably comprise one or more of the regions described above with respect to the hexon protein deletions. For instance, preferably the one or more regions into which the spacer region is inserted or which the spacer region replaces comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48, and equivalents and conservative variations of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID N0:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48.
Similarly, the spacer region itself (i.e., both for insertion as well as replacement) preferably comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID N0:12, SEQ ID N0:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48, and equivalents and conservative variations of SEQ ID NO: 6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48.
The fiber protein also preferably is altered in a similar fashion as described for modification of hexon protein to escape antibodies directed in particular against wild-type adenovirus fiber protein. Fiber protein sequences and methods of modifying fiber protein are known to those skilled in the art (see, e.g., Xia et al . , supra; Novelli et al . , Virology, 185, 365-376 (1991)) . The fiber manipulations can be carried out in the absence of, or along with, modifications to the adenovirus hexon protein. In particular, preferably the fiber protein can be replaced in its entirety, or in part, with sequences of a fiber protein from a different serotype of adenovirus. Also, preferably, deletions can be made of fiber sites that constitute an epitope for a neutralizing antibody, and/or insertions can be made at the site to destroy the ability of the protein to interact with the antibody.
Nucleic Acid Encoding The Chimeric Adenovirus Coat Protein
Preferably the chimeric adenovirus coat protein (particularly the chimeric adenovirus hexon or fiber protein) comprises a nonnative amino acid sequence wherein the alteration is made at the level of DNA. Thus, the invention preferably provides an isolated and purified nucleic acid encoding a chimeric adenovirus coat protein. Desirably, the invention provides an isolated and purified nucleic acid encoding a chimeric adenovirus hexon protein as defined herein, wherein the nucleic acid sequence comprises a deletion of a region (or a plurality of such deletions) that encodes from about 1 to about 750 amino acids of the wild-type adenovirus coat protein, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted comprises a smaller region that encodes less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids. In particular, optimally the deletion (e.g., of an adenoviral hexon protein) comprises the entirety of the 11 and/or 12 loop, or a sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO:47, or a sequence comprising the corresponding region from Adl, Ad3, Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra.
The invention also preferably provides an isolated and purified nucleic acid encoding a chimeric adenovirus hexon protein as defined herein, wherein the nucleic acid sequence comprises a deletion of one or more sequences selected from the group consisting of equivalents and conservatively modified variants of sequences that encode the entirety of the II and/or 12 loop, or SEQ ID NO: 5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID N0:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID N0:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO: 7, or a sequence comprising the corresponding region from Adl, Ad3, Ad6, Ad7 , Ad8, Adll, Adl2, Adl4, Adl6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al . , supra ■
With respect to the nucleic acid sequence, an "equivalent" is a variation on the nucleic acid sequence such as can occur in different strains of adenovirus, and which either does or does not result in a variation at the amino acid level. Failure to result in variation at the amino acid level can be due, for instance, to degeneracy in the triplet code. A "conservatively modified variant" is a variation on the nucleic acid sequence that results in one or more conservative amino acid substitutions . In comparison, a "nonconservatively modified variant" is a variation on the nucleic acid sequence that results in one or more nonconservative amino acid substitutions .
In another preferred embodiment, the invention provides an isolated and purified nucleic acid encoding a chimeric adenovirus coat protein wherein the nucleic acid sequence further comprises a replacement of the deleted region (or a plurality of such replacements) with a spacer nucleic acid region (i.e., the nucleic acid sequence that encodes the aforementioned "spacer region") that encodes from about 1 to about 750 amino acids of the wild-type adenovirus coat protein, preferably from about 1 to about 500 amino acids, and optimally from about 1 to about 300 amino acids. It also is desirable that the region deleted and replaced comprises a smaller region that encodes less than about 200 amino acids, preferably less than about 100 amino acids, and optimally less than about 50 amino acids. Preferably, the spacer nucleic acid region comprises a FLAG octapeptide-encoding sequence [SEQ ID NO:49], and equivalents and conservatively modified variants of SEQ ID NO: 49. Similarly, a spacer nucleic acid region can be employed that substitutes one or more coat protein encoding regions (particularly a hexon protein encoding region) of a particular adenoviral serotype with a coat protein encoding region (particularly a hexon protein encoding region) of another adenoviral serotype. Thus, preferably a spacer nucleic acid region present in a chimeric adenoviral hexon protein is selected from the group consisting of sequences that encode the entirety of the 11 and/or 12 loop, or SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO:47, or a sequence comprising the corresponding region from Adl, Ad3, Ad6, Ad7, Ad8, Adll, Adl2, Adl4, Adl 6, Ad21, Ad34, Ad35, Ad40, Ad41, Ad48, BAV3, or MAVl, especially as reported in Crawford-Miksza et al., supra, and equivalents and conservatively modified variants of these sequences.
As described above with respect to the chimeric adenovirus coat protein, the spacer nucleic acid region (or a plurality thereof) simply can be incorporated into the coat protein in the absence of any deletions . These manipulations can be carried out so as to produce the above-described chimeric adenovirus coat protein.
The means of making such a chimeric adenoviral coat protein (i.e., by introducing conservative or nonconservative variations at either the level of DNA or protein) are known in the art, are described in the Examples which follow, and also can be accomplished by means of various commercially available kits and vectors (e.g., New England Biolabs, Inc., Beverly, MA; Clontech, Palo Alto, CA; Stratagene, LaJolla, CA, and the like) . In particular, the ExSite™ PCR-based site-directed mutagenesis kit and the Chameleon™ double-stranded site- directed mutagenesis kit by Stratagene can be employed for introducing such mutations. Moreover, the means of assessing such mutations (e.g., in terms of effect on ability not to be neutralized by antibodies directed against wild-type hexon protein) are described in the Examples herein.
Accordingly, the present invention provides a preferred means of making a chimeric adenoviral coat protein, particularly a chimeric adenoviral hexon protein, which comprises obtaining an adenoviral genome encoding the wild-type adenovirus coat protein (e.g., the wild-type adenovirus hexon protein) , and deleting one or more region (s) of the chimeric adenovirus coat protein (particularly the chimeric adenovirus hexon protein) comprising from about 1 to about 750 amino acids by modifying the corresponding nucleic acid coding sequence. Similarly, the invention provides a method of making a chimeric adenovirus coat protein (particularly a chimeric adenovirus hexon protein) which comprises obtaining an adenoviral genome encoding the wild-type adenovirus coat protein, deleting one or more region (s) of the adenovirus coat protein comprising from about 1 to about 750 amino acids by modifying the corresponding coding sequence, and replacing the deleted region (s) with a spacer region comprising from about 1 to about 300 amino acids by introducing a nucleic acid region (i.e., a "spacer nucleic acid region") that codes for same. Alternately, the spacer region preferably is simply incorporated into the coat protein (particularly the hexon protein) in the absence of any deletion. Optimally the spacer nucleic acid region encodes a nonconservative variation of the amino acid sequence of the wild-type adenovirus coat protein. The size of the DNA used to replace the native coat protein coding sequence may be constrained, for example, by impeded folding of the coat protein or improper assembly of the coat protein into a complex (e.g., penton base/hexon complex) or virion. DNA encoding 150 amino acids or less is particularly preferred for insertion/replacement in the chimeric coat protein gene sequence, and DNA encoding 50 amino acids or less is even more preferred.
Briefly, the method of mutagenesis comprises deleting one or more regions of an adenovirus coat protein, and/or inserting into an adenovirus coat protein one or more regions with a differing amino acid sequence, particularly by manipulating the DNA sequence. Several methods are available for carrying out such manipulations of adenovirus coat protein DNA sequences; these methods further can be used in combination. The method of choice depends on factors known to those skilled in the art, e.g., the size of the DNA region to be manipulated. For instance, convenient restriction sites (which further can be introduced into a sequence) can be used to introduce or remove segments of DNA, or entire genes or coding sequences. Alternately, other methods of mutagenesis involve the hybridization of a mismatched oligonucleotide to a region of single-stranded target DNA, extending the primer, for instance, using T7 DNA polymerase or other such means to produce a double-stranded heteroduplex, and isolating the mutant strand that incorporates the mismatched oligonucleotide from the parental nonmutant strand for use as a template and in further manipulations . The mutant strand can be separated from the parental strand using various selection means known to those skilled in the art (see, e.g., Kunkel et al . , Methods Enzymol. , 204, 125-139 (1991), as well as the underlying methodology employed in the Chameleon™ kit) . Alternately, the parental strand can be selectively degraded, for instance, with use of enzymes that nick the nonmethylated strand of a hemi-methylated DNA molecule (e.g., Jfpall, Mspl , and Sau3AI), and by extending the mutant strand using 5-methyl-dCTP, which renders the strand resistant to cleavage by these enzymes. Along the same lines, an entirely PCR-based approach can be employed for making mutations (e.g., Kunkel, Proc. Natl . Acad. Sci., 82, 488- 492 (1985); Costa et al . , Nucleic Acids Res., 22_, 2423 (1994)), for instance, such as the approach encompassed by the ExSite™ kit. More generally, amino acid substitutions or deletions can be introduced during PCR by incorporating appropriate mismatches in one or both primers. Once the chimeric coat protein sequence has been produced, the nucleic acid fragment encoding the sequence further can be isolated, e.g., by PCR amplification using 5' and 3' primers, or through use of convenient restriction sites.
Vector Comprising a Chimeric Hexon Protein
A "vector" according to the invention is a vehicle for gene transfer as that term is understood by those skilled in the art, and includes viruses, plasmids, and the like. A preferred vector is an adenovirus, particularly a virus of the family Adenoviridae, and desirably of the genus Mastadenovirus (e.g., comprised of mammalian adenoviruses) or Aviadenovirus (e.g., comprised of avian adenoviruses) . Such an adenovirus (or other viral vector) can be transferred by its own means of effecting cell entry (e.g., by receptor-mediated endocytosis) , or can be transferred to a cell like a plasmid, i.e., in the form of its nucleic acid, for instance, by using liposomes to transfer the nucleic acid, or by microinjecting or transforming the DNA into the cell. The nucleic acid vectors that can be employed for gene transfer, particularly the adenoviral nucleic acid vectors, are referred to herein as "transfer vectors". Such nucleic acid vectors also include intermediary plasmid vectors that are employed, e.g., in the construction of adenoviral vectors.
Desirably an adenoviral vector is a serotype group C virus, preferably an Ad2 or Ad5 vector, although any other serotype adenoviral vector (e.g., group A including serotypes 12 and 31, group B including serotypes 3 and 7, group D including serotypes 8 and 30, group E including serotype 4, and group F including serotypes 40 and 41, and other Ad vectors previously described) can be employed. An adenoviral vector employed for gene transfer can be replication competent. Alternately, an adenoviral vector can comprise genetic material with at least one modification therein, which renders the virus replication deficient. The modification to the adenoviral genome can include, but is not limited to, addition of a DNA segment, rearrangement of a DNA segment, deletion of a DNA segment, replacement of a DNA segment, or introduction of a DNA lesion. A DNA segment can be as small as one nucleotide and as large as 36 kilobase pairs (i.e., the approximate size of the adenoviral genome) or, alternately, can equal the maximum amount which can be packaged into an adenoviral virion (i.e., about 38 kb) . Preferred modifications to the group C adenoviral genome include modifications in the El, E2, E3 and/or E4 regions. Similarly, an adenoviral vector can be a cointegrate, i.e., a ligation of adenoviral sequences with other sequences, such as other virus sequences, particularly baculovirus sequences, or plasmid sequences, e.g., so as to comprise a prokaryotic or eukaryotic expression vector.
In terms of an adenoviral vector (particularly a replication deficient adenoviral vector) , such a vector can comprise either complete capsids (i.e., including a viral genome such as an adenoviral genome) or empty capsids (i.e., in which a viral genome is lacking, or is degraded, e.g., by physical or chemical means) . The capsid further can comprise nucleic acid linked to the surface by means known in the art (e.g., Curiel et al . , Human Gene Therapy, 3_, 147-154 (1992) ) or can transfer non-linked nucleic acid, for instance, by adenoviral- mediated uptake of bystander nucleic acid (e.g., PCT International Application WO 95/21259) .
Along the same lines, since methods are available for transferring an adenovirus in the form of its nucleic acid sequence (i.e., DNA), a vector (i.e., a transfer vector) similarly can comprise DNA, in the absence of any associated protein such as capsid protein, and in the absence of any envelope lipid. Inasmuch as techniques are available for making a RNA copy of DNA (e.g., in vi tro transcription) , and inasmuch as RNA viruses also can be employed as vectors or transfer vectors, a transfer vector also can comprise RNA. Thus, according to the invention whereas a vector comprises (and, further, may encode) a chimeric adenoviral coat protein, a transfer vector typically encodes a chimeric adenoviral coat protein (particularly a chimeric adenoviral hexon and/or fiber protein) .
Based on this, the invention provides an adenoviral vector that comprises a chimeric coat protein (particularly a chimeric hexon and/or fiber protein) according to the invention. Preferably such a vector comprises a chimeric coat protein (particularly a chimeric adenovirus hexon protein and/or chimeric adenovirus fiber protein) as described above. Alternately, preferably the vector lacks wild-type fiber protein, e.g., the vector encodes a truncated or non-functional fiber protein, or fails to translate fiber protein. Such fiber mutations and the means of introducing fiber mutations are known to those skilled in the art (see, e.g., Falgout et al . , J. Virol., 6_, 622-625 (1988)).
Of course, the chimeric adenoviral coat proteins include coat proteins in which the native (i.e., wild- type) hexon and/or fiber protein of an adenoviral vector is replaced by a hexon and or fiber amino acid sequence of a different adenoviral serotype such that the resultant adenoviral vector has a decreased ability or inability to be recognized by neutralizing antibodies directed against the corresponding wild-type coat protein. This replacement can comprise the entirety of the hexon and/or fiber amino acid sequence, or only a portion, as described above. Both proteins can be manipulated (e.g., in a single adenovirus), or only a single chimeric adenovirus coat protein can be employed, with the remaining coat proteins being wild-type.
A vector according to the invention (including a transfer vector) preferably comprises additional sequences and mutations, e.g., some that can occur within the coat protein coding sequence itself. In particular, a vector according to the invention further preferably comprises a nucleic acid encoding a passenger gene or passenger coding sequence. A "nucleic acid" is a polynucleotide (i.e., DNA or RNA) . A "gene" is any nucleic acid sequence coding for a protein or an RNA molecule. Whereas a gene comprises coding sequences plus any non-coding sequences, a "coding sequence" does not include any non-coding (e.g., regulatory) DNA. A "passenger gene" or "passenger coding sequence" is any gene which is not typically present in and is subcloned into a vector (e.g., a transfer vector) according to the present invention, and which upon introduction into a host cell is accompanied by a discernible change in the intracellular environment (e.g., by an increased level of deoxyribonucleic acid (DNA) , ribonucleic acid (RNA) , peptide or protein, or by an altered rate of production or degradation thereof) . A "gene product" is either an as yet untranslated RNA molecule transcribed from a given gene or coding sequence (e.g., mRNA or antisense RNA) or the polypeptide chain (i.e., protein or peptide) translated from the mRNA molecule transcribed from the given gene or coding sequence. A gene or coding sequence is "recombinant" if the sequence of bases along the molecule has been altered from the sequence in which the gene or coding sequence is typically found in nature, or if the sequence of bases is not typically found in nature. According to this invention, a gene or coding sequence can be naturally occurring or wholly or partially synthetically made, can comprise genomic or complementary DNA (cDNA) sequences, and can be provided in the form of either DNA or RNA.
Non-coding sequences or regulatory sequences include promoter sequences. A "promoter" is a DNA sequence that directs the binding of RNA polymerase and thereby promotes RNA synthesis. "Enhancers" are cis-acting elements of DNA that stimulate or inhibit transcription of adjacent genes. An enhancer that inhibits transcription is also termed a "silencer". Enhancers differ from DNA-binding sites for sequence-specific DNA binding proteins found only in the promoter (which also are termed "promoter elements") in that enhancers can function in either orientation, and over distances of up to several kilobase pairs, even from a position downstream of a transcribed region. According to the invention, a coding sequence is "operably linked" to a promoter (e.g., when both the coding sequence and the promoter constitute a passenger gene) when the promoter is capable of directing transcription of that coding sequence .
Accordingly, a "passenger gene" can be any gene, and desirably either is a therapeutic gene or a reporter gene. Preferably a passenger gene is capable of being expressed in a cell in which the vector has been internalized. For instance, the passenger gene can comprise a reporter gene, or a nucleic acid sequence which encodes a protein that can be detected in a cell in some fashion. The passenger gene also can comprise a therapeutic gene, for instance, a therapeutic gene which exerts its effect at the level of RNA or protein. Similarly, a protein encoded by a transferred therapeutic gene can be employed in the treatment of an inherited disease, such as, e.g., the cystic fibrosis transmembrane conductance regulator cDNA for the treatment of cystic fibrosis. The protein encoded by the therapeutic gene can exert its therapeutic effect by resulting in cell killing. For instance, expression of the gene in itself may lead to cell killing, as with expression of the diphtheria toxin A gene, or the expression of the gene may render cells selectively sensitive to the killing action of certain drugs, e.g., expression of the HSV thymidine kinase gene renders cells sensitive to antiviral compounds including acyclovir, gancyclovir and FIAU ( 1- (2-deoxy-2-fluoro-b-D- arabinofuranosil) -5-iodouracil) . Moreover, the therapeutic gene can exert its effect at the level of RNA, for instance, by encoding an antisense message or ribozyme, by affecting splicing or 3' processing (e.g., polyadenylation) , or by encoding a protein which acts by affecting the level of expression of another gene within the cell (i.e., where gene expression is broadly considered to include all steps from initiation of transcription through production of a processed protein) , perhaps, among other things, by mediating an altered rate of mRNA accumulation, an alteration of mRNA transport, and/or a change in post-transcriptional regulation. Accordingly, the use of the term "therapeutic gene" is intended to encompass these and any other embodiments of that which is more commonly referred to as gene therapy and is known to those of skill in the art. Similarly, the recombinant adenovirus can be used for gene therapy or to study the effects of expression of the gene (e.g., a reporter gene) in a given cell or tissue in vi tro or in vivo, or for diagnostic purposes.
Also, a passenger coding sequence can be employed in the vector. Such a coding sequence can be employed for a variety of purposes even though a functional gene product may not be translated from the vector sequence. For instance, the coding sequence can be used as a substrate for a recombination reaction, e.g., to recombine the sequence with the host cell genome or a vector resident in the cell. The coding sequence also can be an "anticoding sequence," e.g., as appropriate for antisense approaches. Other means of using the coding sequence will be known to one skilled in the art.
The present invention thus provides recombinant adenoviruses comprising a chimeric hexon protein and/or a chimeric fiber protein, and which preferably additionally comprise a passenger gene or genes capable of being expressed in a particular cell. The recombinant adenoviruses can be generated by use of a vector, specifically, a transfer vector, and preferably a viral (especially an adenoviral) or plasmid transfer vector, in accordance with the present invention. Such a transfer vector preferably comprises a chimeric adenoviral hexon and/or fiber gene sequence as previously described.
Similarly, the means of constructing such a transfer vector are known to those skilled in the art. For instance, a chimeric adenovirus coat protein gene sequence can simply be ligated into the vector using convenient restriction sites. Alternately, a wild-type adenovirus gene sequence can be mutagenized to create the chimeric coat protein sequence following its subcloning into a vector. Similarly, a chimeric coat protein gene sequence can be moved via standard molecular genetic techniques from a transfer vector into baculovirus or a suitable prokaryotic or eukaryotic expression vector (e.g., a viral or plasmid vector) for expression and evaluation of penton base binding, and other biochemical characteristics.
Accordingly, the present invention also provides recombinant baculoviral and prokaryotic and eukaryotic expression vectors comprising an aforementioned chimeric adenoviral coat protein gene sequence, which, along with the nucleic acid form of the adenoviral vector (i.e., an adenoviral transfer vector) are "transfer vectors" as defined herein. By moving the chimeric gene from an adenoviral vector to baculovirus or a prokaryotic or eukaryotic expression vector, high protein expression is achievable (approximately 5-50% of the total protein being the chimeric protein) .
Similarly, adenoviral vectors (e.g., virions or virus particles) are produced using transfer vectors. For instance, an adenoviral vector comprising a chimeric coat protein according to the invention can be constructed by introducing into a cell, e.g., a 293 cell, a vector comprising sequences from the adenoviral left arm, and a vector comprising sequences from the adenoviral right arm, wherein there is a region of overlap between the sequences. As described in the Examples which follow, this methodology results in recombination between the sequences, generating a vector that comprises a portion of each of the vectors, particularly the region comprising the chimeric coat protein sequences.
The present invention thus preferably also provides a method of constructing an adenoviral vector that has a decreased ability or inability to be recognized by a neutralizing antibody directed against wild-type adenovirus hexon protein and/or fiber protein. This method comprises replacing a coat protein of the vector (i.e., a wild-type adenovirus hexon and/or fiber protein) with the corresponding chimeric adenovirus coat protein according to the invention to produce a recombinant adenoviral vector.
The coat protein chimera-containing particles are produced in standard cell lines, e.g., those currently used for adenoviral vectors . Deletion mutants lacking the fiber gene, or possessing shortened versions of the fiber protein, similarly can be employed in vector construction, e.g., H2dI802, H2dl807, H2dll021 (Falgout et al . , supra) , as can other fiber mutants. The fiberless particles have been shown to be stable and capable of binding and infecting cells (Falgout et al . , supra) .
Illustrative Uses and Benefits
The present invention provides a chimeric coat protein that has a decreased ability or inability to be recognized by a neutralizing antibody directed against the corresponding wild-type coat protein, as well as vectors (including transfer vectors) comprising same. The chimeric coat protein (such as a chimeric hexon and/or fiber protein) has multiple uses, e.g., as a tool for studies in vitro of capsid structure and assembly, and capsomere binding to other proteins.
A vector (e.g., a transfer vector) comprising a chimeric coat protein can be used in strain generation, for instance, in generation of recombinant strains of adenovirus. Similarly, such a vector, particularly an adenoviral vector, can be used in gene therapy. Specifically, a vector of the present invention can be used to treat any one of a number of diseases by delivering to targeted cells corrective DNA, i.e., DNA encoding a function that is either absent or impaired, or a discrete killing agent, e.g., DNA encoding a cytotoxin that, for instance, is active only intracellularly . Diseases that are candidates for such treatment include, but are not limited to, cancer, e.g., melanoma, glioma or lung cancers; genetic disorders, e.g., cystic fibrosis, hemophilia or muscular dystrophy; pathogenic infections, e.g., human immunodeficiency virus, tuberculosis or hepatitis; heart disease, e.g., preventing restenosis following angioplasty or promoting angiogenesis to reperfuse necrotic tissue; and autoimmune disorders, e.g., Crohn ' s disease, colitis or rheumatoid arthritis. In particular, gene therapy can be carried out in the treatment of diseases, disorders, or conditions that require repeat administration of the corrective DNA and/or the adenoviral vector, and thus for which current adenoviral-mediated approaches to gene therapy are less than optimal .
Moreover, such a vector, particularly an adenoviral vector, can be used to deliver material to a cell not as a method of gene therapy, but for diagnostic or research purposes. In particular, a vector comprising a chimeric adenovirus coat protein according to the invention can be employed to deliver a gene either in vitro or in vivo, for research and/or diagnostic purposes.
For instance, instead of transferring a so-called therapeutic gene, a reporter gene or some type of marker gene can be transferred instead. Marker genes and reporter genes are of use, for instance, in cell differentiation and cell fate studies, as well as potentially for diagnostic purposes. Moreover, a standard reporter gene such as a β-galactosidase reporter gene, a gene encoding green fluorescent protein (GFP) , or a β- glucuronidase gene can be used in vivo, e.g., as a means of assay in a living host, or, for instance, as a means of targeted cell ablation (see, e.g., Minden et al . , BioTechniques, 20, 122-129 (1996); Youvan, Science, 268, 264 (1995); U.S. Patent 5,432,081; Deonarain et al . , Br . J. Cancer, 1_, 786-794 (1994)).
Similarly, it may be desirable to transfer a gene to use a host essentially as a means of production in vivo of a particular protein. Along these lines, transgenic animals have been- employed, for instance, for the production of recombinant polypeptides in the milk of transgenic bovine species (e.g., PCT International Application WO 93/25567) . The use of an adenovirus according to the invention for gene transfer conducted for protein production in vivo further is advantageous in that such use should result in a reduced (if not absent) immune response as compared with the use of a wild-type adenovirus vector. Other "non-therapeutic" reasons for gene transfer include the study of human diseases using an animal model (e.g., use of transgenic mice and other transgenic animals including p53 tumor suppressor gene knockouts for tumorigenic studies, use of a transgenic model for impaired glucose tolerance and human Alzheimer's amyloid precursor protein models for the study of glucose metabolism and for the pathogenesis of Alzheimer's disease, respectively, etc.).
Furthermore, an adenoviral vector comprising a chimeric adenovirus coat protein and employed as described above is advantageous in that it can be isolated and purified by conventional means. For instance, it is likely that special cell lines will not need to be made in order to propagate adenoviruses comprising the chimeric coat proteins .
These aforementioned illustrative uses and recitation of benefits are by no means comprehensive, and it is intended that the present invention encompass such further uses which necessarily flow from, but are not explicitly recited, in the disclosure herein. Means of Administration
The vectors and transfer vectors of the present invention can be employed to contact cells either in vi tro or in vivo . According to the invention "contacting" comprises any means by which a vector is introduced intracellularly; the method is not dependent on any particular means of introduction and is not to be so construed. Means of introduction are well known to those skilled in the art, and also are exemplified herein.
Accordingly, introduction can be effected, for instance, either in vi tro (e.g., in an ex vivo type method of gene therapy or in tissue culture studies) or in vivo by methods that include, but are not limited to, electroporation, transformation, transduction, conjugation, triparental mating, (co-) transfection, (co- ) infection, high velocity bombardment with DNA-coated microprojectiles, incubation with calcium phosphate-DNA precipitate, direct microinjection into single cells, and the like. Similarly, the vectors can be introduced by means of membrane fusion using cationic lipids, e.g., liposomes. Such liposomes are commercially available (e.g., Lipofectin®, Lipofectamine™, and the like, supplied by Life Technologies, Gibco BRL, Gaithersburg, MD) . Moreover, liposomes having increased transfer capacity and/or reduced toxicity in vivo (see, e.g., PCT International Application WO 95/21259 and references reviewed therein) can be employed in the present invention. Other methods also are available and are known to those skilled in the art.
According to the invention, a "host" encompasses any host into which a vector of the invention can be introduced, and thus encompasses an animal, including, but not limited to, an amphibian, bird, insect, reptile, or mammal. Optimally a host is a mammal, for instance, a rodent, primate (such as chimpanzee, monkey, ape, gorilla, orangutan, or gibbon), feline, canine, ungulate (such as ruminant or swine), as well as, in particular, a human.
Similarly, a "cell" encompasses any cell (or collection of cells) from a host into which an adenoviral vector can be introduced, e.g., preferably an epithelial cell. Any suitable organs or tissues or component cells can be targeted for vector delivery. Preferably, the organs/tissues/cells employed are of the circulatory system (e.g., heart, blood vessels or blood), respiratory system (e.g., nose, pharynx, larynx, trachea, bronchi, bronchioles, lungs), gastrointestinal system (e.g., mouth, pharynx, esophagus, stomach, intestines, salivary glands, pancreas, liver, gallbladder), urinary system (e.g., kidneys, ureters, urinary bladder, urethra), nervous system (e.g. brain and spinal cord, or special sense organs such as the eye) and integumentary system (e.g., skin) . Even more preferably the cells being targeted are selected from the group consisting of heart, blood vessel, lung, liver, gallbladder, urinary bladder, and eye cells.
Thus, the present invention preferably also provides a method of genetically modifying a cell. This method preferably comprises contacting a cell with a vector comprising a chimeric adenovirus hexon protein and/or a chimeric adenovirus fiber protein, wherein desirably the vector is an adenovirus vector. The method preferably results in the production of a host cell comprising a vector according to the invention.
Moreover, the method of the invention of genetically modifying a cell can be employed in gene therapy, or for administration for diagnosis or study. The application of this method in vivo optimally comprises administering to a patient in need of gene therapy (e.g., a patient suffering from a disease, condition or disorder) a therapeutically effective amount of a recombinant adenovirus vector according to the invention. This method preferably can be employed as part of an ongoing gene therapy regimen, e.g., wherein the vector (e.g., a recombinant adenovirus vector) comprising the chimeric adenovirus coat protein is administered following (e.g., after from about 1 week to about 2 months) administration of a therapeutically effective amount of a vector comprising either the corresponding wild-type coat protein or a coat protein of a different adenoviral serotype. Alternately, the vector comprising the chimeric adenovirus coat protein can be employed as an initial attempt at gene delivery.
One skilled in the art will appreciate that suitable methods of administering a vector (particularly an adenoviral vector) of the present invention to an animal for purposes of gene therapy (see, for example, Rosenfeld et al. (1991), supra; Jaffe et al., Clin. Res., 39 (2) , 302A (1991); Rosenfeld et al., Clin. Res., 39(2) , 311A (1991a); Berkner, supra) , chemotherapy, vaccination, diagnosis, and/or further study are available. Although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. For instance, local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, peritoneal, subcutaneous, intradermal, as well as topical administration. Clinical trials regarding use of gene therapy vectors in vivo are ongoing. The methodology employed for such clinical trials as well as further technologies known to those skilled in the art can be used to administer the vector of the present invention for the purpose of research, diagnosis and/or gene therapy. Pharmaceutically acceptable excipients also are well- known to those who are skilled in the art, and are readily available. The choice of excipient will be determined in part by the particular method used to administer the recombinant vector. Accordingly, there is a wide variety of suitable formulations for use in the context of the present invention. The following methods and excipients are merely exemplary and are in no way limiting.
Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules; (c) suspensions in an appropriate liquid; and (d) suitable emulsions. Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
A vector of the present invention (including an adenoviral vector and a transfer vector) , alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They may also be formulated as pharmaceuticals for non-pressured preparations such as in a nebulizer or an atomizer .
Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
Additionally, a vector of the present invention can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
The dose administered to an animal, particularly a human, in the context of the present invention will vary with the gene of interest, the composition employed, the method of administration, the particular site and organism undergoing administration, and the reason for the administration (e.g., gene therapy, diagnosis, means of producing a protein, further study, etc) . Generally, the "effective amount" of the composition is such as to produce the desired effect in a host which can be monitored using several end-points known to those skilled in the art. For example, one desired effect might comprise effective nucleic acid transfer to a host cell. Such transfer can be monitored in terms of a therapeutic effect (e.g., alleviation of some symptom associated with the disease or syndrome being treated) , or by further evidence of the transferred gene or coding sequence or its expression within the host (e.g., using the polymerase chain reaction, Northern or Southern hybridizations, or transcription assays to detect the nucleic acid in host cells, or using immunoblot analysis, antibody-mediated detection, or particularized assays to detect protein or polypeptide encoded by the transferred nucleic acid, or impacted in level or function due to such transfer) . One such particularized assay described in the Examples which follow includes an assay for expression of a chloramphenicol acetyl transferase reporter gene.
Generally, to ensure effective transfer of the vectors of the present invention, it is preferable that from about 1 to about 5,000 copies of the vector be employed per cell to be contacted, based on an approximate number of cells to be contacted in view of the given route of administration. It is even more preferable that from about 1 to about 300 plaque forming units (pfu) enter each cell. However, this is just a general guideline which by no means precludes use of a higher or lower amount of a component, as might be warranted in a particular application, either in vi tro or in vivo . For example, the actual dose and schedule can vary depending on whether the composition is administered in combination with other pharmaceutical compositions, or depending on interindividual differences in pharmacokinetics, drug disposition, and metabolism. Similarly, amounts can vary in in vi tro applications depending on the particular cell type utilized or the means by which the vector is transferred. One skilled in the art easily can make any necessary adjustments in accordance with the necessities of the particular situation.
The following examples further illustrate the present invention and, of course, should not be construed as in any way limiting its scope.
Example 1
This example describes experiments investigating adenoviral anti-vector neutralizing immunity.
To clarify the phenomenon of neutralizing immunity, an animal having circulating antibodies to one adenoviral vector type received intratracheal administration of another serotype adenoviral vector, and gene expression commanded by the second vector was monitored. Specifically, either an Ad4 or Ad5 wild-type vector was administered to the lungs of Sprague-Dawley rats. Ten days later, an Ad5 reporter vector was administered to the lungs of the same animals. This reporter vector, which is referred to herein as the "pure 5" vector, comprises an El" E3~ type 5 adenoviral vector which expresses the chloramphenicol acetyl transferase (CAT) gene driven by the cytomegalovirus early/intermediate promoter/enhancer (CMV) (i.e., AdCMVCATgD described in Kass-Eisler et al., Proc. Natl. Acad. Sci., 5, 11498-11502 (1993)).
About twenty-four hours following administration of the "pure 5" vector, CAT activity was measured in homogenized lung tissue using a CAT assay as previously described (Kass Eisler et al . (1993), supra) . CAT activity was monitored at various times thereafter up to 10 days following introduction of the "pure 5" vector. CAT activity was determined relative to the "pure 5" vector administered to naive animals (i.e., expression measured under this condition was considered 100%). The results of these studies are set out in Table 1, and are further reported in Mastrangeli et al . , Human Gene Therapy, 1 , 79-87 (1996) .
Figure imgf000048_0001
These results confirm that in the presence of neutralizing antibodies elicited against one adenoviral group (e.g., against group E, serotype 4), it is possible to efficiently transfer and express a gene in vivo using an adenoviral vector derived from another group (e.g., derived from group C, serotype 5) . Neutralizing immunity evoked against one serotype group does not protect against infection by another group of adenovirus. These data support the paradigm of alternating adenoviral vectors derived from different subgroups as a strategy to circumvent anti-adenoviral humoral immunity.
Example 2
The predominant epitopes that evoke neutralizing immunity are located on the fiber and hexon, but mainly on hexon. Based on this, the effect of switching the fiber protein was investigated. A vector was constructed that was identical to the "pure 5" vector except that the fiber gene was switched from a serotype 5, group C fiber to a serotype 7, group B fiber. The resultant vector is referred to herein as the "5 base/7 fiber" vector.
The Ad5/Ad7 fiber construct was generated as shown in Figure 1. An approximately 2.7 kb (Ad5 28689-31317 bp) fragment in pAd70-100 was replaced with a Pad linker (pAd70-100dlE3.Pac) . A BamHI linker was inserted at a Muni site as indicated in Figure 2 to produce pAd70- 100dlE3.Pac.Bam. A PCR-amplified Pacl-BamHI fragment of approximately 1.1 kb containing the Ad7 fiber gene was inserted into pAd70-100dlE3. Pac .Bam to produce pAd70- 100dlE3.fiber7.
In order to assess the ability of the Ad5 virus with Ad7 fiber to infect cells in vi tro and in vivo, reporter gene assays were performed. A replication-defective recombinant adenoviral reporter vector designated AdCMV- CATNeo was used in the reporter gene assay. The reporter vector consists of the adenoviral origin of replication and viral packaging sequences, a combination of strong eukaryotic promoter (cytomegalovirus or CMV-1) and splicing elements, the bacterial chloramphenicol acetyl transferase (CAT) gene sequence, the mouse βmaj-globin poly (A) site, the neomycin gene sequence (Neo) , and sufficient adenoviral DNA to allow for overlap recombination .
The reporter vector was used to generate AdCMV- CATNeo, AdCMV-CATNeo-dlE3 (AdCMV-CATNeo + pAd70-100dlE3) and AdCMV-CATNeo-dlE3-Fiber7 (AdCMV-CATNeo + pAd70- 1001E3. Fiber7 ) viruses. Each virus was grown in large scale, i.e., a one liter suspension of human embryonic kidney 293 cells, to yield virus at a concentration of 1012 particles/ml. A549 cells were infected with an estimated 100, 300 or 1,000 particles/cell of one of the three viruses. After 48 hours, the cells were harvested and lysates were prepared as described in Kass-Eisler et al . (1993) , supra . Using 50 μl of each lysate, CAT assays were performed and acetylated chloramphenicol products were separated by thin layer chromatography using chloroform:methanol (95:5). The results of the assays confirm that each virus was able to infect cells and express gene products at appropriate levels. Accordingly, the virus in which the native fiber was replaced with a nonnative fiber could infect cells and express genes like the parental virus .
Following this study, adult Sprague-Dawley rats were infected with 108 viral particles by direct cardiac injection as described in Kass-Eisler et al . (1993), supra . Five days later, the rats were sacrificed, cardiac lysates were prepared, and CAT assays were performed. The amount of the CAT gene product produced was compared between the dlE3 and dlE3-Fiber7 viruses. Results indicated that both viruses were able to infect cells in vivo . The replacement of the wild-type Ad5 fiber gene with that of Ad7 did not impair the ability of the virus to infect cells. Accordingly, the virus in which the native fiber was replaced with a nonnative fiber could also infect cells and express genes like the parental virus in vivo . These results support the utility of adenovirus with chimeric fiber in the context of gene therapy.
Example 3
This example describes the effect on neutralizing immunity of switching the fiber protein of an adenovirus from one serotype to another.
The "pure 5" and "5 base/7 fiber" vectors described in the preceding Example were administered to Sprague- Dawley rats which either were naive or pre-immunized against wild-type Ad5. For these experiments, wild-type Ad5 or wild-type Ad7 (6 x 109 particles in phosphate buffered saline (PBS) ) was administered intraperitoneally as a primary inoculation. Seventeen days later, serum samples were taken, and about 6 x 109 particles in about 50 μl of PBS was injected. At about 120 hours following injection the animals were sacrificed, serum and heart tissue were harvested, and heart tissue was processed for CAT assays as previously described (Kass-Eisler et al . (1993), supra) . CAT assays also were performed on heart lysates of rat hearts infected with the "pure 5" vector or "5 base/7 fiber" vector alone.
Administration of either vector to naive animals resulted in comparable levels of CAT in heart tissue. In comparison, administration of either the "pure 5" vector or the "5 base/7 fiber" vector to the animals that were pre-immunized against the "pure 5" vector resulted in a reduction of CAT levels by more than two orders of magnitude as compared with mock-infected controls. These and further results are reported in Gall et al . , J. Virol., 70, 2116-2163 (1996).
These results confirm that switching the fiber from that of adenoviral serotype 5 group C vector to that of an adenoviral serotype 7 group B vector by itself is insufficient to allow the vector to escape neutralizing antibodies generated against an adenoviral vector comprising Ad5 fiber. These results imply that antibodies against adenoviral structures other than fiber also are important in the process of neutralizing immunity. Furthermore, whereas switching the fiber serotype to another serotype may be insufficient in and of itself to allow an adenovirus to escape immune detection, such switching when done in combination with removal of other epitopes may be desirable, for instance, to reduce an immune response. Example 4
This example describes the construction of adenovirus vectors wherein the neutralizing immunity-evoking epitopes have been modified. In particular, this example describes vectors comprising chimeric adenoviral hexon protein, wherein the hexon neutralizing immunity-evoking epitopes are modified.
The results of the prior example indicate that it is possible to develop vectors for repeat administration in gene therapy from non-group C adenovirus, thus circumventing pre-existing neutralizing immunity. As another strategy, the dominant neutralizing immunity- evoking epitopes on existing group C vectors can be modified to render the vectors less susceptible (or "stealth") to the existing neutralizing immunity. For instance, adenoviral type 5-based El" E3~ CAT-expressing vectors can be constructed that have the same genetic composition as the "pure 5" and "5 base/7 fiber" vectors described above, except for possessing a gene encoding a chimeric hexon that is not recognized by pre-existing anti-type 5 neutralizing immunity.
To derive the vectors, the chimeric hexon gene present in the "pure 5" parental vector can be modified, in particular, II and/or 12 can be altered. The hexon modifications that can be made on the "pure 5" CAT vector, or other adenoviral vector (such as any other adenoviral serotype vector), include, but are not limited to: (1) hexon with 11 deleted in its entirety; (2) hexon with 12 deleted in its entirety; (3) hexon with both 11 and 12 deleted; (4) hexon with any one or more of HVRl, HVR2, HVR3, HVR4, HVR5 , HVR6, or HVR7 , deleted; (5) -(8) hexon with a FLAG octamer epitope (i.e., Asp Tyr Lys Asp Asp Asp Asp Lys [SEQ ID NO: 50]; Hopp et al . , Biotechnology, 6_, 1205-1210 (1988)) substituted for 11, 12, or both 11 and 12, or any one or more of HVRl, HVR2, HVR3, HVR , HVR5, HVR6 or HVR7; (9) -(12) hexon with a FLAG octamer epitope [SEQ ID NO:50] inserted into 11, 12, or both II and 12; (13) -(16) hexon with comparable epitopes from Ad7 (group B) (GenBank® Data Bank Accession Number x76551 for Ad7 hexon, and Number M73260 for Ad5 hexon) or Ad2, or any other adenoviral serotype, substituted for 11, 12, both 11 and 12, respectively, or for any one or more of HVRl, HVR2, HVR3, HVR , HVR5, HVR6, or HVR7 ; (17) -(20) hexon with comparable epitopes from Ad7 (group B) (GenBank® Data Bank Accession Number x76551 for Ad7 hexon, and Number M73260 for Ad5 hexon) or Ad2, or any other adenoviral serotype, inserted into 11, 12, both 11 and 12, respectively, or any one or more of HVRl, HVR2, HVR3, HVR4, HVR5, HVR6, or HVR7 ; and (21) complete substitution of the hexon from Ad2 or another adenoviral serotype, for the Ad5 hexon. The use of the FLAG octamer epitope provides a sequence for incorporation in the chimeric hexon protein that is different from the Ad5 hexon loop sequences, and also provides a positive control using available specific anti-FLAG antibodies (Hopp et al . , supra) .
These chimeric hexon proteins (and vectors containing them) can be made in several steps. To modify the hexon in the "pure 5" vector, a viral or plasmid vector can be constructed to contain the hexon type 5 coding sequence in a cassette that can be easily modified. The hexon is read off the 1 strand of the L3 transcription unit, i.e., map units 51.6 to 59.7, comprising a region of about 2.9 kb. The two other transcripts that also are encoded by L3 -- i.e., polypeptide VI and a 23 kDa protein -- do not overlap the hexon coding sequence. Moreover, there are no other coding sequences on the r strand that would be altered by the modification of the hexon coding sequence.
Thus, all the modifications of the type 5 hexon can be made using a "hexon 5 cassette" comprised of an approximate 6.7 kb Sfil-Sfil fragment of the "pure 5" CAT vector. Sfil cuts Ad5 into 3 fragments, the center 6.7 kb fragment (i.e., comprising about 16,282 to 22,992 base pairs, as identified by agarose gel electrophoresis) of which contains all of the L3 region plus some overlap. The "hexon 5 cassette" can be subcloned into a commercially available vector having restriction sites and the like making the vector easily manipulable in terms of modification and recovery of subcloned sequences. One such vector appropriate for subcloning is either the SK or KS version of the pBlueScript® phagemid (Stratagene, LaJolla, CA) .
The "hexon 5 cassette" can be mutagenized to generate site-specific mutations in the cloned DNA segment. Several methods are available for carrying out site- specific mutagenesis. The 11 and 12 deletions, insertions, or replacements (or deletions, insertions, or replacements in HVRl, HVR2, HVR3, HVR4 , HVR5, HVR6, or HVR7 regions contained therein) can be made by deleting the relevant sequences using restriction enzymes that cut uniquely within the vector inserts, or other similar means, e.g., by ligating in an end-polished, or otherwise modified, PCR product. Alternately, the hexon sequence contained in the hexon 5 cassette can be modified, e.g., using single-stranded mutagenesis in M13mp8 or some other convenient vector, and using appropriate oligonueleotides encompassing the flanking sequences for identification of plaques as described by Crompton et al . , supra. Alternately, a commercially available kit such as the ExSite™ PCR-based site-directed mutagenesis kit and the Chameleon™ double-stranded site-directed mutagenesis kit by Stratagene can be used to introduce insertions, point mutations, or deletions into the chimeric hexon sequence without any need for subcloning into an M13, or other special vector. Similarly, the FLAG octapeptide sequence (Hopp et al . , supra) can be introduced into the vectors (i.e., in the presence or absence of any deletion) by inserting the relevant 24 base pair sequence (GAY TAY AAR GAY GAY GAY GAY AAR [SEQ ID NO:50], wherein Y is C or T/ϋ, and R is A or G) ) . The replacement of Ad5 hexon loop epitopes with comparable sequences of Ad7 , Ad2, or any other adenoviral serotype, or an incorporation of these sequences in the absence of any deletion, can be accomplished by using unique restriction sites, or using one of the aforementioned means of mutagenesis. This usefully creates new serotypes of adenoviral vectors. For example, The replacement of the wildtype hexon protein of Ad5 with the chimeric Ad5 hexon comprising Ad7 hexon loops 1 and 2 gives rise to an adenoviral vector that is effectively neutralized by Ad7 neutralizing antibodies (i.e., neutralizing antibodies raised in response to Ad7 innoculation of a naive animal), but not by Ad5 neutralizing antibodies.
Moreover, both hypervariable loops 1 and 2 can be deleted from a serotype 5 or another serotype adenoviral vector. Adenoviral vectors and there genomes comprising these deletions are useful as a starting point to create other adenoviral vectors having loop replacements, as a tool for studying hexon structure-function relationships, and under some circumstances as a gene transfer vector with limited vulnerability to the adaptive immune system.
Example 5
This example describes the method of replacing the hexon protein of one serotype adenoviral vector with the hexon protein of another serotype adenoviral vector to generate a recombinant adenovirus . As representative of this method, the hexon protein of an Ad5 vector was replaced with the hexon protein of an Ad2 vector. This example also describes the method of incorporating the chimeric hexon proteins of the preceding Example into a vector to make a recombinant adenovirus.
Using standard molecular biology techniques, the Ad5 hexon gene open reading frame (ORF) was replaced with the Ad2 hexon gene ORF in such a fashion so as to maintain the proper Ad5 sequences upstream and downstream of the hexon gene. Adenoviral vectors comprising modified or chimeric hexon proteins can be constructed by homologous recombination using standard techniques and human embryonic kidney 293 cells (see, e.g., Rosenfeld et al . (1991), supra; Rosenfeld et al . (1992), supra) . For instance, map units 0 to 57.3 of dlAd5NCAT (Gall et al . , supra) can be isolated by Bsu36l digestion, and map units 58.4 to 100 of dlAd5NCAT can be isolated by Drdl digestion. These DNA fragments can be transfected into 293 cells along with pH5-2.
A neutralizing antibody directed against the parental vector can be employed to facilitate the generation of hexon replacement constructs. For example, when replacing the loop 1 and loop 2 regions of an Ad5 vector with Ad7 loop sequences, anti-Ad5 neutralizing polyclonal or monoclonal antibodies (directed against the loops 1 and 2 of Ad5 hexon) can be added to a the medium of cells in which the chimeric vector is being propagated. The presence of the Ad5 neutralizing antibodies substantially blocks the propagation of the undesired wildtype Ad5 vector (s), while the chimeric vector is unaffected. Furthermore, the recombinant vectors comprising a chimeric hexon ORF can be generated by homologous recombination using a plasmid that carries a marker gene, such as Green Fluorescent Protein (GFP) , adjacent to the chimeric or novel hexon ORF (e.g., between the fiber and hexon genes) . In this way, genomes that could harbor the chimeric hexon gene should also harbor the marker gene. The marker gene would then be expressed as a late protein, so that cells that potentially comprise the desired adenoviral genome can be easily identified.
Similarly, vectors (particularly adenoviral vectors) can be constructed that have the aforementioned hexon modifications, and which have further modifications, for instance, in the adenoviral fiber coding sequences. This can be accomplished by making the hexon modifications described above, and using different parental plasmids for homologous recombination, such as parental plasmids comprising mutations in fiber coding sequences. In particular, the "5 base/7 fiber" vector can be employed as a starting vector for vector construction.
All of the viral vectors prepared according to this example can be plaque-purified, amplified, and further purified using standard methods (Rosenfeld et al . (1991), supra; Rosenfeld et al . (1992), supra) .
Example 6
This example describes a characterization of the activity in vi tro and in vivo of the vectors described in the preceding Examples.
Each of the viruses prepared as described in the preceding Examples can be evaluated in vi tro and in vivo using standard methods as previously described (e.g., Kass-Eisler et al . , supra) , and as set forth herein. In particular, for the in vitro studies, the various vectors along with control vectors (e.g., the "pure 5" and "5 base/7 fiber" vectors, and the Ad5 wild-type vector) can be added to human lung carcinoma A549 cells alone, or in the presence of dilutions of serum from hosts infected with Ad5, Ad7, "pure 5" CAT vector, or "5 base/7 fiber" CAT vector, or anti-FLAG epitope serum. The cells are then evaluated for CAT activity to determine the ability of antibodies present in the serum to block gene expression .
The in vivo studies can be carried out in Sprague- Dawley rats. The Sprague-Dawley rat as opposed to the mouse or cotton rat is preferred for these experiments since the rat is non-permissive, and the wild-type adenovirus cannot replicate in this host. Accordingly, immunizations can be carried out using wild-type viruses (e.g., wild-type Ad5 or Ad7 ) , the "pure 5" CAT vector, and the "5 base/7 fiber" CAT vector by intravenous administration (e.g., Kass-Eisler et al . , supra) . At various times ranging from about one to about four weeks later, the vector of interest can be administered intravenously or directly into the airways of the host. Whereas intravenous administration allows an assessment of the "worst case scenario" (i.e., wherein the vector is in immediate contact with the circulating humoral immune system, and thus the strongest immune response is to be expected) , introduction in the airways of the host allows an evaluation of a compartmentalized and mucosal humoral immune response.
CAT activity can be quantified as previously described in all the relevant organs, e.g., liver, heart, and lung for intravenous administration, and lung only for respiratory administration. Appropriate standards can-be used to compensate for variations in organ expression of CAT activity (see e.g., Kass-Eisler et al . , Gene Therapy, 2_ 395-402 (1994)) . The in vitro and in vivo results can be compared and assessed using standard statistical methods .
All of the references cited herein, including the GenBank® Data Bank sequence information, are hereby incorporated in their entireties by reference. While this invention has been described with emphasis upon preferred embodiments, it will be obvious to those of ordinary skill in the art that the preferred embodiments can be varied. It is intended that the invention can be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications encompassed within the spirit and scope of the appended claims .
SEQUENCE LISTING
(1) GENERAL INFORMATION: (i) APPLICANT:
NAME: CORNELL RESEARCH FOUNDATION, INC.
STREET: 20 Thornwood Drive, Suite 105
CITY: Ithaca
STATE: New York
COUNTRY: US
POSTAL CODE (ZIP) : 14850
TELEPHONE: 607-257-1081
TELEFAX: 607-257-1015
NAME: GENVEC, INC.
STREET: 12111 Parklawn Drive
CITY: Rockville
STATE: Maryland
COUNTRY : US
POSTAL CODE (ZIP) : 20852
TELEPHONE: 301-816-0396
TELEFAX: 301-816-0085
NAME: CRYSTAL, RONALD G.
STREET: 13712 Canal Vista Court
CITY: Potomac
STATE: Maryland
COUNTRY: US
POSTAL CODE (ZIP) : 20854
NAME: FALCK-PEDERSEN, ERIK
STREET: 8714 Buena Vista Drive
CITY: Dobbs Ferry
STATE: New York
COUNTRY: US
POSTAL CODE (ZIP) : 10522
NAME: GALL, JASON
STREET: 420 E. 70th Street, #10F
CITY: New York
STATE: New York
COUNTRY: US
POSTAL CODE (ZIP) : 10021
NAME: KOVESDI, IMRE
STREET: 7713 Warbler Lane
CITY: Rockville
STATE: Maryland
COUNTRY: US
POSTAL CODE (ZIP) : 20855
NAME: WICKHAM, THOMAS J.
STREET: 2106 Hutchinson Grove Court
CITY: Falls Church
STATE: Virginia
COUNTRY: US
POSTAL CODE (ZIP) : 22043
(ii) TITLE OF INVENTION: CHIMERIC ADENOVIRAL COAT PROTEIN AND METHODS OF USING SAME
(iii) NUMBER OF SEQUENCES: 56
(iv) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentln Release #1.0, Version #1.30 (EPO)
(vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 8-816346
(B) FILING DATE: 13-MAR-1997
(2) INFORMATION FOR SEQ ID NO : 1 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2907 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
ATG GCT ACC CCT TCG ATG ATG CCG CAG TGG TCT TAC ATG CAC ATC TCG 48 Met Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His lie Ser 1 5 10 15
GGC CAG GAC GCC TCG GAG TAC CTG AGC CCC GGG CTG GTG CAG TTT GCC 96 Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
CGC GCC ACC GAG ACG TAC TTC AGC CTG AAT AAC AAG TTT AGA AAC CCC 144 Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
ACG GTG GCA CCT ACG CAC GAC GTA ACC ACA GAC CGG TCC CAG CGT TTG 192 Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
ACG CTG CGG TTC ATC CCT GTG GAC CGC GAG GAT ACC GCG TAC TCG TAC 240 Thr Leu Arg Phe lie Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75 80
AAA GCG CGG TTC ACC CTG GCT GTG GGT GAC AAC CGT GTG CTT GAT ATG 288 Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 85 90 95
GCT TCC ACG TAC TTT GAC ATC CGC GGC GTG CTG GAC AGG GGG CCT ACT 336 Ala Ser Thr Tyr Phe Asp lie Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
TTT AAG CCC TAC TCC GGC ACT GCC TAC AAC GCT CTA GCT CCC AAG GGC 384 Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
GCT CCT AAC TCC TGT GAG TGG GAA CAA ACC GAA GAT AGC GGC CGG GCA 432 Ala Pro Asn Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala 130 135 140
GTT GCC GAG GAT GAA GAA GAG GAA GAT GAA GAT GAA GAA GAG GAA GAA 480 Val Ala Glu Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu 145 150 155 160
GAA GAG CAA AAC GCT CGA GAT CAG GCT ACT AAG AAA ACA CAT GTC TAT 528 Glu Glu Gin Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr 165 170 175 GCC CAG GCT CCT TTG TCT GGA GAA ACA ATT ACA AAA AGC GGG CTA CAA 576 Ala Gin Ala Pro Leu Ser Gly Glu Thr lie Thr Lys Ser Gly Leu Gin 180 185 190
ATA GGA TCA GAC AAT GCA GAA ACA CAA GCT AAA CCT GTA TAC GCA GAT 624 lie Gly Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp 195 200 205
CCT TCC TAT CAA CCA GAA CCT CAA ATT GGC GAA TCT CAG TGG AAC GAA 672 Pro Ser Tyr Gin Pro Glu Pro Gin lie Gly Glu Ser Gin Trp Asn Glu 210 215 220
GCT GAT GCT AAT GCG GCA GGA GGG AGA GTG CTT AAA AAA ACA ACT CCC 720 Ala Asp Ala Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro 225 230 235 240
ATG AAA CCA TGC TAT GGA TCT TAT GCC AGG CCT ACA AAT CCT TTT GGT 768 Met Lys Pro Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly 245 250 255
GGT CAA TCC GTT CTG GTT CCG GAT GAA AAA GGG GTG CCT CTT CCA AAG 816 Gly Gin Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys 260 265 270
GTT GAC TTG CAA TTC TTC TCA AAT ACT ACC TCT TTG AAC GAC CGG CAA 864 Val Asp Leu Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin 275 280 285
GGC AAT GCT ACT AAA CCA AAA GTG GTT TTG TAC AGT GAA GAT GTA AAT 912 Gly Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn 290 295 300
ATG GAA ACC CCA GAC ACA CAT CTG TCT TAC AAA CCT GGA AAA GGT GAT 960 Met Glu Thr Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp 305 310 315 320
GAA AAT TCT AAA GCT ATG TTG GGT CAA CAA TCT ATG CCA AAC AGA CCC 1008 Glu Asn Ser Lys Ala Met Leu Gly Gin Gin Ser Met Pro Asn Arg Pro 325 330 335
AAT TAC ATT GCT TTC AGG GAC AAT TTT ATT GGC CTA ATG TAT TAT AAC 1056 Asn Tyr lie Ala Phe Arg Asp Asn Phe lie Gly Leu Met Tyr Tyr Asn 340 345 350
AGC ACT GGC AAC ATG GGT GTT CTT GCT GGT CAG GCA TCG CAG CTA AAT 1104 Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala Ser Gin Leu Asn 355 360 365
GCC GTG GTA GAT TTG CAA GAC AGA AAC ACA GAG CTG TCC TAT CAA CTC 1152 Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu Ser Tyr Gin Leu 370 375 380
TTG CTT GAT TCC ATA GGT GAT AGA ACC AGA TAT TTT TCT ATG TGG AAT 1200 Leu Leu Asp Ser lie Gly Asp Arg Thr Arg Tyr Phe Ser Met Trp Asn 385 390 395 400
CAG GCT GTA GAC AGC TAT GAT CCA GAT GTT AGA ATC ATT GAA AAC CAT 1248 Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg lie lie Glu Asn His 405 410 415
GGA ACT GAG GAT GAA TTG CCA AAT TAT TGT TTT CCT CTT GGG GGT ATT 1296 Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro Leu Gly Gly lie 420 425 430 GGG GTA ACT GAC ACC TAT CAA GCT ATT AAG GCT AAT GGC AAT GGC TCA 1344 Gly Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser 435 440 445
GGC GAT AAT GGA GAT ACT ACA TGG ACA AAA GAT GAA ACT TTT GCA ACA 1392 Gly Asp Asn Gly Asp Thr Thr Trp Thr Lys Asp Glu Thr Phe Ala Thr 450 455 460
CGT AAT GAA ATA GGA GTG GGT AAC AAC TTT GCC ATG GAA ATT AAC CTA 1440 Arg Asn Glu He Gly Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 465 470 475 480
AAT GCC AAC CTA TGG AGA AAT TTC CTT TAC TCC AAT ATT GCG CTG TAC 1488 Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 485 490 495
CTG CCA GAC AAG CTA AAA TAC AAC CCC ACC AAT GTG GAA ATA TCT GAC 1536 Leu Pro Asp Lys Leu Lys Tyr Asn Pro Thr Asn Val Glu He Ser Asp 500 505 510
AAC CCC AAC ACC TAC GAC TAC ATG AAC AAG CGA GTG GTG GCT CCC GGG 1584 Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 515 520 525
CTT GTA GAC TGC TAC ATT AAC CTT GGG GCG CGC TGG TCT CTG GAC TAC 1632 Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 530 535 540
ATG GAC AAC GTT AAT CCC TTT AAC CAC CAC CGC AAT GCG GGC CTC CGT 1680 Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 545 550 555 560
TAT CGC TCC ATG TTG TTG GGA AAC GGC CGC TAC GTG CCC TTT CAC ATT 1728 Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 565 570 575
CAG GTG CCC CAA AAG TTT TTT GCC ATT AAA AAC CTC CTC CTC CTG CCA 1776 Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 580 585 590
GGC TCA TAT ACA TAT GAA TGG AAC TTC AGG AAG GAT GTT AAC ATG GTT 1824 Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 595 600 605
CTG CAG AGC TCT CTG GGA AAC GAT CTT AGA GTT GAC GGG GCT AGC ATT 1872 Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 610 615 620
AAG TTT GAC AGC ATT TGT CTT TAC GCC ACC TTC TTC CCC ATG GCC CAC 1920 Lys Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His 625 630 635 640
AAC ACG GCC TCC ACG CTG GAA GCC ATG CTC AGA AAT GAC ACC AAC GAC 1968 Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 645 650 655
CAG TCC TTT AAT GAC TAC CTT TCC GCC GCC AAC ATG CTA TAC CCC ATA 2016 Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 660 665 670
CCC GCC AAC GCC ACC AAC GTG CCC ATC TCC ATC CCA TCG CGC AAC TGG 2064 Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 675 680 685 GCA GCA TTT CGC GGT TGG GCC TTC ACA CGC TTG AAG ACA AAG GAA ACC 2112 Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 690 695 700
CCT TCC CTG GGA TCA GGC TAC GAC CCT TAC TAC ACC TAC TCT GGC TCC 2160 Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 705 710 715 720
ATA CCA TAC CTT GAC GGA ACC TTC TAT CTT AAT CAC ACC TTT AAG AAG 2208 He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 725 730 735
GTG GCC ATT ACC TTT GAC TCT TCT GTT AGC TGG CCG GGC AAC GAC CGC 2256 Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 740 745 750
CTG CTT ACT CCC AAT GAG TTT GAG ATT AAA CGC TCA GTT GAC GGG GAG 2304 Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 755 760 765
GGC TAC AAC GTA GCT CAG TGC AAC ATG ACC AAG GAC TGG TTC CTG GTG 2352 Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 770 775 780
CAG ATG TTG GCC AAC TAC AAT ATT GGC TAC CAG GGC TTC TAC ATT CCA 2400 Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 785 790 795 800
GAA AGC TAC AAG GAC CGC ATG TAC TCG TTC TTC AGA AAC TTC CAG CCC 2448 Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 805 810 815
ATG AGC CGG CAA GTG GTT GAC GAT ACT AAA TAC AAG GAG TAT CAG CAG 2496 Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Glu Tyr Gin Gin 820 825 830
GTT GGA ATT CTT CAC CAG CAT AAC AAC TCA GGA TTC GTA GGC TAC CTC 2544 Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 835 840 845
GCT CCC ACC ATG CGC GAG GGA CAG GCT TAC CCC GCC AAC GTG CCC TAC 2592 Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Val Pro Tyr 850 855 860
CCA CTA ATA GGC AAA ACC GCG GTT GAC AGT ATT ACC CAG AAA AAG TTT 2640 Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 865 870 875 880
CTT TGC GAT CGC ACC CTT TGG CGC ATC CCA TTC TCC AGT AAC TTT ATG 2688 Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 885 890 895
TCC ATG GGC GCA CTC ACA GAC CTG GGC CAA AAC CTT CTC TAC GCC AAC 2736 Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 900 905 910
TCC GCC CAC GCG CTA GAC ATG ACT TTT GAG GTG GAT CCC ATG GAC GAG 2784 Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 915 920 925
CCC ACC CTT CTT TAT GTT TTG TTT GAA GTC TTT GAC GTG GTC CGT GTG 2832 Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 930 935 940 CAC CAG CCG CAC CGC GGC GTC ATC GAG ACC GTG TAC CTG CGC ACG CCC 2880 His Gin Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 945 950 955 960
TTC TCG GCC GGC AAC GCC ACA ACA TAA 2907
Phe Ser Ala Gly Asn Ala Thr Thr 965
(2) INFORMATION FOR SEQ ID NO : 2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 968 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser 1 5 10 15
Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
Thr Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75 80
Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 85 90 95
Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
Ala Pro Asn Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala 130 135 140
Val Ala Glu Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu 145 150 155 160
Glu Glu Gin Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr 165 170 175
Ala Gin Ala Pro Leu Ser Gly Glu Thr He Thr Lys Ser Gly Leu Gin 180 185 190
He Gly Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp 195 200 205
Pro Ser Tyr Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp Asn Glu 210 215 220
Ala Asp Ala Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro 225 230 235 240 Met Lys Pro Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly 245 250 255
Gly Gin Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys 260 265 270
Val Asp Leu Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin 275 280 285
Gly Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn 290 295 300
Met Glu Thr Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp 305 310 315 320
Glu Asn Ser Lys Ala Met Leu Gly Gin Gin Ser Met Pro Asn Arg Pro 325 330 335
Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu Met Tyr Tyr Asn 340 345 350
Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala Ser Gin Leu Asn 355 360 365
Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu Ser Tyr Gin Leu 370 375 380
Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe Ser Met Trp Asn 385 390 395 400
Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He He Glu Asn His 405 410 415
Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro Leu Gly Gly He 420 425 430
Gly Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser 435 440 445
Gly Asp Asn Gly Asp Thr Thr Trp Thr Lys Asp Glu Thr Phe Ala Thr 450 455 460
Arg Asn Glu He Gly Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 465 470 475 480
Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 485 490 495
Leu Pro Asp Lys Leu Lys Tyr Asn Pro Thr Asn Val Glu He Ser Asp 500 505 510
Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 515 520 525
Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 530 535 540
Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 545 550 555 560
Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 565 570 575 Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 580 585 590
Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 595 600 605
Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 610 615 620
Lys Phe Asp Ser He C s Leu Tyr Ala Thr Phe Phe Pro Met Ala His 625 630 635 640
Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 645 650 655
Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 660 665 670
Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 675 680 685
Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 690 695 700
Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 705 710 715 720
He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 725 730 735
Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 740 745 750
Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 755 760 765
Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 770 775 780
Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 785 790 795 800
Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 805 810 815
Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Glu Tyr Gin Gin 820 825 830
Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 835 840 845
Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Val Pro Tyr 850 855 860
Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 865 870 875 880
Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 885 890 895
Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 900 905 910 Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 915 920 925
Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 930 935 940
His Gin Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 945 950 955 960
Phe Ser Ala Gly Asn Ala Thr Thr
(2) INFORMATION FOR SEQ ID NO : 3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2858 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE
(A) NAME/KEY: misc_feature
(B) LOCATION: 951, 952
(D) OTHER INFORMATION: /note="Xaa can be either Gin, His, or Thr"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
ATG GCT ACC CCT TCG ATG ATG CCG CAG TGG TCT TAC ATG CAC ATC TCG 48 Met Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser 1 5 10 15
GGC CAG GAC GCC TCG GAG TAC CTG AGC CCC GGG CTG GTG CAG TTT GCC 96 Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
CGC GCC ACC GAG ACG TAC TTC AGC CTG AAT AAC AAG TTT AGA AAC CCC 144 Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
ACG GTG GCG CCT ACG CAC GAC GTG ACC ACA GAC CGG TCC CAG CGT TTG 192 Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
ACG CTG CGG TTC ATC CCT GTG GAC CGT GAG GAT ACT GCG TAC TCG TAC . 240 Thr Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75 80
AAG GCG CGG TTC ACC CTA GCT GTG GGT GAT AAC CGT GTG CTG GAC ATG 288 Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 85 90 95
GCT TCC ACG TAC TTT GAC ATC CGC GGC GTG CTG GAC AGG GGC CCT ACT 336 Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
TTT AAG CCC TAC TCT GGC ACT GCC TAC AAC GCC CTG GCT CCC AAG GGT 384 Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
GCC CCA AAT CCT TGC GAA TGG GAT GAA GCT GCT ACT GCT CTT GAA ATA 432 Ala Pro Asn Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He 130 135 140 AAC CTA GAA GAA GAG GAC GAT GAC AAC GAA GAC GAA GTA GAC GAG CAA 480 Asn Leu Glu Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin 145 150 155 160
GCT GAG CAG CAA AAA ACT CAC GTA TTT GGG CAG GCG CCT TAT TCT GGT 528 Ala Glu Gin Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly 165 170 175
ATA AAT ATT ACA AAG GAG GGT ATT CAA ATA GGT GTC GAA GGT CAA ACA 576 He Asn He Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr 180 185 190
CCT AAA TAT GCC GAT AAA ACA TTT CAA CCT GAA CCT CAA ATA GGA GAA 624 Pro Lys Tyr Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu 195 200 205
TCT CAG TGG TAC GAA ACT GAA ATT AAT CAT GCA GCT GGG AGA GTC CTT 672 Ser Gin Trp Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu 210 215 220
AAA AAG ACT ACC CCA ATG AAA CCA TGT TAC GGT TCA TAT GCA AAA CCC 720 Lys Lys Thr Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro 225 230 235 240
ACA AAT GAA AAT GGA GGG CAA GGC ATT CTT GTA AAG CAA CAA AAT GGA 768 Thr Asn Glu Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly 245 250 255
AAG CTA GAA AGT CAA GTG GAA ATG CAA TTT TTC TCA ACT ACT GAG GCG 816 Lys Leu Glu Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala 260 265 270
ACC GCA GGC AAT GGT GAT AAC TTG ACT CCT AAA GTG GTA TTG TAC AGT 864 Thr Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser 275 280 285
GAA GAT GTA GAT ATA GAA ACC CCA GAC ACT CAT ATT TCT TAC ATG CCC 912 Glu Asp Val Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro 290 295 300
ACT ATT AAG GAA GGT AAC TCA CGA GAA CTA ATG GGC CAA CAA TCT ATG 960 Thr He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met 305 310 315 320
CCC AAC AGG CCT AAT TAC ATT GCT TTT AGG GAC AAT TTT ATT GGT CTA 1008 Pro Asn Arg Pro Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu 325 330 335
ATG TAT TAC AAC AGC ACG GGT AAT ATG GGT GTT CTG GCG GGC CAA GCA 1056 Met Tyr Tyr Asn Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala 340 345 350
TCG CAG TTG AAT GCT GTT GTA GAT TTG CAA GAC AGA AAC ACA GAG CTT 1104 Ser Gin Leu Asn Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu 355 360 365
TCA TAC CAG CTT TTG CTT GAT TCC ATT GGT GAT AGA ACC AGG TAC TTT 1152 Ser Tyr Gin Leu Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe 370 375 380
TCT ATG TGG AAT CAG GCT GTT GAC AGC TAT GAT CCA GAT GTT AGA ATT 1200 Ser Met Trp Asn Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He 385 390 395 400 ATT GAA AAT CAT GGA ACT GAA GAT GAA CTT CCA AAT TAC TGC TTT CCA 1248 He Glu Asn His Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro 405 410 415
CTG GGA GGT GTG ATT AAT ACA GAG ACT CTT ACC AAG GTA AAA CCT AAA 1296 Leu Gly Gly Val He Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys 420 425 430
ACA GGT CAG GAA AAT GGA TGG GAA AAA GAT GCT ACA GAA TTT TCA GAT 1344 Thr Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp 435 440 445
AAA AAT GAA ATA AGA GTT GGA AAT AAT TTT GCC ATG GAA ATC AAT CTA 1392 Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 450 455 460
AAT GCC AAC CTG TGG AGA AAT TTC CTG TAC TCC AAC ATA GCG CTG TAT 1440 Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 465 470 475 480
TTG CCC GAC AAG CTA AAG TAC AGT CCT TCC AAC GTA AAA ATT TCT GAT 1488 Leu Pro Asp Lys Leu Lys Tyr Ser Pro Ser Asn Val Lys He Ser Asp 485 490 495
AAC CCA AAC ACC TAC GAC TAC ATG AAC AAG CGA GTG GTG GCT CCC GGG 1536 Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 500 505 510
TTA GTG GAC TGC TAC ATT AAC CTT GGA GCA CGC TGG TCC CTT GAC TAT 1584 Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 515 520 525
ATG GAC AAC GTC AAC CCA TTT AAC CAC CAC CGC AAT GCT GGC CTG CGC 1632 Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 530 535 540
TAC CGC TCA ATG TTG CTG GGC AAT GGT CGC TAT GTG CCC TTC CAC ATC 1680 Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 545 550 555 560
CAG GTG CCT CAG AAG TTC TTT GCC ATT AAA AAC CTC CTT CTC CTG CCG 1728 Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 565 570 575
GGC TCA TAC ACC TAC GAG TGG AAC TTC AGG AAG GAT GTT AAC ATG GTT 1776 Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 580 585 590
CTG CAG AGC TCC CTA GGA AAT GAC CTA AGG GTT GAC GGA GCC AGC ATT 1824 Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 595 600 605
AAG TTT GAT AGC ATT TGC CTT TAC GCC ACC TTC TTC CCC ATG GCC CAC 1872 Lys Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His 610 615 620
AAC ACC GCC TCC ACG CTT GAG GCC ATG CTT AGA AAC GAC ACC AAC GAC 1920 Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 625 630 635 640
CAG TCC TTT AAC GAC TAT CTC TCC GCC GCC AAC ATG CTC TAC CCT ATA 1968 Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 645 650 655 CCC GCC AAC GCT ACC AAC GTG CCC ATA TCC ATC CCC TCC CGC AAC TGG 2016 Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 660 665 670
GCG GCT TTC CGC GGC TGG GCC TTC ACG CGC CTT AAG ACT AAG GAA ACC 2064 Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 675 680 685
CCA TCA CTG GGC TCG GGC TAC GAC CCT TAT TAC ACC TAC TCT GGC TCT 2112 Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 690 695 700
ATA CCC TAC CTA GAT GGA ACC TTT TAC CTC AAC CAC ACC TTT AAG AAG 2160 He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 705 710 715 720
GTG GCC ATT ACC TTT GAC TCT TCT GTC AGC TGG CCT GGC AAT GAC CGC 2208 Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 725 730 735
CTG CTT ACC CCC AAC GAG TTT GAA ATT AAG CGC TCA GTT GAC GGG GAG 2256 Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 740 745 750
GGT TAC AAC GTT GCC CAG TGT AAC ATG ACC AAA GAC TGG TTC CTG GTA 2304 Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 755 760 765
CAA ATG CTA GCT AAC TAC AAC ATT GGC TAC CAG GGC TTC TAT ATC CCA 2352 Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 770 775 780
GAG AGC TAC AAG GAC CGC ATG TAC TCC TTC TTT AGA AAC TTC CAG CCC 2400 Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 785 790 795 800
ATG AGC CGT CAG GTG GTG GAT GAT ACT AAA TAC AAG GAC TAC CAA CAG 2448 Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Asp Tyr Gin Gin 805 810 815
GTG GGC ATC CTA CAC CAA CAC AAC AAC TCT GGA TTT GTT GGC TAC CTT 2496 Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 820 825 830
GCC CCC ACC ATG CGC GAA GGA CAG GCC TAC CCT GCT AAC TTC CCC TAT 2544 Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Phe Pro Tyr 835 840 845
CCG CTT ATA GGC AAG ACC GCA GTT GAC AGC ATT ACC CAG AAA AAG TTT 2592 Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 850 855 860
CTT TGC GAT CGC ACC CTT TGG CGC ATC CCA TTC TCC AGT AAC TTT ATG 2640 Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 865 870 875 880
TCC ATG GGC GCA CTC ACA GAC CTG GGC CAA AAC CTT CTC TAC GCC AAC 2688 Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 885 890 895
TCC GCC CAC GCG CTA GAC ATG ACT TTT GAG GTG GAT CCC ATG GAC GAG 2736 Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 900 905 910 CCC ACC CTT CTT TAT GTT TTG TTT GAA GTC TTT GAC GTG GTC CGT GTG 2784 Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 915 920 925
CAC CGG CCG CAC CGC GGC GTC ATC GAA ACC GTG TAC CTG CGC ACG CCC 2832 His Arg Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 930 935 940
TTC TCG GCC GGC AAC GCA HHH HHH HH 2858
Phe Ser Ala Gly Asn Ala Xaa Xaa 945 950
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 952 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(ix) FEATURE
(A) NAME/KEY: misc_feature
(B) LOCATION: 951,952
(D) OTHER INFORMATION: /note= "Xaa can be either Gin, His, or Thr"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
Met Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser 1 5 10 15
Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
Thr Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75 80
Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 85 90 95
Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
Ala Pro Asn Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He 130 135 140
Asn Leu Glu Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin 145 150 155 160
Ala Glu Gin Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly 165 170 175
He Asn He Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr 180 185 190 Pro Lys Tyr Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu 195 200 205
Ser Gin Trp Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu 210 215 220
Lys Lys Thr Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro 225 230 235 240
Thr Asn Glu Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly 245 250 255
Lys Leu Glu Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala 260 265 270
Thr Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser 275 280 285
Glu Asp Val Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro 290 295 300
Thr He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met 305 310 315 320
Pro Asn Arg Pro Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu 325 330 335
Met Tyr Tyr Asn Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala 340 345 350
Ser Gin Leu Asn Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu 355 360 365
Ser Tyr Gin Leu Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe 370 375 380
Ser Met Trp Asn Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He 385 390 395 400
He Glu Asn His Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro 405 410 415
Leu Gly Gly Val He Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys 420 425 430
Thr Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp 435 440 445
Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 450 455 460
Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 465 470 475 480
Leu Pro Asp Lys Leu Lys Tyr Ser Pro Ser Asn Val Lys He Ser Asp 485 490 495
Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 500 505 510
Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 515 520 525 Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 530 535 540
Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 545 550 555 560
Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 565 570 575
Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 580 585 590
Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 595 600 605
Lys Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His 610 615 620
Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 625 630 635 640
Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 645 650 655
Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 660 665 670
Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 675 680 685
Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 690 695 700
He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 705 710 715 720
Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 725 730 735
Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 740 745 750
Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 755 760 765
Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 770 775 780
Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 785 790 795 800
Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Asp Tyr Gin Gin 805 810 815
Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 820 825 830
Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Phe Pro Tyr 835 840 845
Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 850 855 860 Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 865 870 875 880
Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 885 890 895
Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 900 905 910
Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 915 920 925
His Arg Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 930 935 940
Phe Ser Ala Gly Asn Ala Xaa Xaa 945 950
(2) INFORMATION FOR SEQ ID NO : 5 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 603 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5:
TCC TGT GAG TGG GAA CAA ACC GAA GAT AGC GGC CGG GCA GTT GCC GAG 48 Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala Val Ala Glu 1 5 10 15
GAT GAA GAA GAG GAA GAT GAA GAT GAA GAA GAG GAA GAA GAA GAG CAA 96 Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu Glu Glu Gin 20 25 30
AAC GCT CGA GAT CAG GCT ACT AAG AAA ACA CAT GTC TAT GCC CAG GCT 144 Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr Ala Gin Ala 35 40 45
CCT TTG TCT GGA GAA ACA ATT ACA AAA AGC GGG CTA CAA ATA GGA TCA 192 Pro Leu Ser Gly Glu Thr He Thr Lys Ser Gly Leu Gin He Gly Ser 50 55 60
GAC AAT GCA GAA ACA CAA GCT AAA CCT GTA TAC GCA GAT CCT TCC TAT 240 Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp Pro Ser Tyr 65 70 75 80
CAA CCA GAA CCT CAA ATT GGC GAA TCT CAG TGG AAC GAA GCT GAT GCT 288 Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp Asn Glu Ala Asp Ala 85 90 95
AAT GCG GCA GGA GGG AGA GTG CTT AAA AAA ACA ACT CCC ATG AAA CCA 336 Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro Met Lys Pro 100 105 110
TGC TAT GGA TCT TAT GCC AGG CCT ACA AAT CCT TTT GGT GGT CAA TCC 384 Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly Gly Gin Ser 115 120 125 GTT CTG GTT CCG GAT GAA AAA GGG GTG CCT CTT CCA AAG GTT GAC TTG 432 Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys Val Asp Leu 130 135 140
CAA TTC TTC TCA AAT ACT ACC TCT TTG AAC GAC CGG CAA GGC AAT GCT 480 Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin Gly Asn Ala 145 150 155 160
ACT AAA CCA AAA GTG GTT TTG TAC AGT GAA GAT GTA AAT ATG GAA ACC 528 Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn Met Glu Thr 165 170 175
CCA GAC ACA CAT CTG TCT TAC AAA CCT GGA AAA GGT GAT GAA AAT TCT 576 Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp Glu Asn Ser 180 185 190
AAA GCT ATG TTG GGT CAA CAA TCT ATG 603
Lys Ala Met Leu Gly Gin Gin Ser Met 195 200
(2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 201 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:
Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala Val Ala Glu 1 5 10 15
Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu Glu Glu Gin 20 25 30
Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr Ala Gin Ala 35 40 45
Pro Leu Ser Gly Glu Thr He Thr Lys Ser Gly Leu Gin He Gly Ser 50 55 60
Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp Pro Ser Tyr 65 70 75 80
Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp Asn Glu Ala Asp Ala 85 90 95
Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro Met Lys Pro 100 105 110
Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly Gly Gin Ser 115 120 125
Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys Val Asp Leu 130 135 140
Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin Gly Asn Ala 145 150 155 160
Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn Met Glu Thr 165 170 175 Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp Glu Asn Ser 180 185 190
Lys Ala Met Leu Gly Gin Gin Ser Met 195 200
(2) INFORMATION FOR SEQ ID NO: 7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 567 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7:
CCT TGC GAA TGG GAT GAA GCT GCT ACT GCT CTT GAA ATA AAC CTA GAA 48 Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He Asn Leu Glu 1 5 10 15
GAA GAG GAC GAT GAC AAC GAA GAC GAA GTA GAC GAG CAA GCT GAG CAG 96 Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin Ala Glu Gin 20 25 30
CAA AAA ACT CAC GTA TTT GGG CAG GCG CCT TAT TCT GGT ATA AAT ATT 144 Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly He Asn He 35 40 45
ACA AAG GAG GGT ATT CAA ATA GGT GTC GAA GGT CAA ACA CCT AAA TAT 192 Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr Pro Lys Tyr 50 55 60
GCC GAT AAA ACA TTT CAA CCT GAA CCT CAA ATA GGA GAA TCT CAG TGG 240 Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp 65 70 75 80
TAC GAA ACT GAA ATT AAT CAT GCA GCT GGG AGA GTC CTT AAA AAG ACT 288 Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu Lys Lys Thr 85 90 95
ACC CCA ATG AAA CCA TGT TAC GGT TCA TAT GCA AAA CCC ACA AAT GAA 336 Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro Thr Asn Glu 100 105 110
AAT GGA GGG CAA GGC ATT CTT GTA AAG CAA CAA AAT GGA AAG CTA GAA 384 Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly Lys Leu Glu 115 120 125
AGT CAA GTG GAA ATG CAA TTT TTC TCA ACT ACT GAG GCG ACC GCA GGC 432 Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala Thr Ala Gly 130 135 140
AAT GGT GAT AAC TTG ACT CCT AAA GTG GTA TTG TAC AGT GAA GAT GTA 480 Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser Glu Asp Val 145 150 155 160
GAT ATA GAA ACC CCA GAC ACT CAT ATT TCT TAC ATG CCC ACT ATT AAG 528 Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro Thr He Lys 165 170 175
GAA GGT AAC TCA CGA GAA CTA ATG GGC CAA CAA TCT ATG 567 Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met 180 185
(2) INFORMATION FOR SEQ ID NO : 8 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 189 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He Asn Leu Glu 1 5 10 15
Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin Ala Glu Gin 20 25 30
Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly He Asn He 35 40 45
Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr Pro Lys Tyr 50 55 60
Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp 65 70 75 80
Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu Lys Lys Thr 85 90 95
Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro Thr Asn Glu 100 105 110
Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly Lys Leu Glu 115 120 125
Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala Thr Ala Gly 130 135 140
Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser Glu Asp Val 145 150 155 160
Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro Thr He Lys 165 170 175
Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met 180 185
(2) INFORMATION FOR SEQ ID NO : 9 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 153 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9: ACC GAA GAT AGC GGC CGG GCA GTT GCC GAG GAT GAA GAA GAG GAA GAT Thr Glu Asp Ser Gly Arg Ala Val Ala Glu Asp Glu Glu Glu Glu Asp 1 5 10 15
GAA GAT GAA GAA GAG GAA GAA GAA GAG CAA AAC GCT CGA GAT CAG GCT 96 Glu Asp Glu Glu Glu Glu Glu Glu Glu Gin Asn Ala Arg Asp Gin Ala 20 25 30
ACT AAG AAA ACA CAT GTC TAT GCC CAG GCT CCT TTG TCT GGA GAA ACA 144 Thr Lys Lys Thr His Val Tyr Ala Gin Ala Pro Leu Ser Gly Glu Thr 35 40 45
ATT ACA AAA 153
He Thr Lys 50
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 51 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
Thr Glu Asp Ser Gly Arg Ala Val Ala Glu Asp Glu Glu Glu Glu Asp 1 5 10 15
Glu Asp Glu Glu Glu Glu Glu Glu Glu Gin Asn Ala Arg Asp Gin Ala 20 25 30
Thr Lys Lys Thr His Val Tyr Ala Gin Ala Pro Leu Ser Gly Glu Thr 35 40 45
He Thr Lys 50
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 135 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
GCT GCT ACT GCT CTT GAA ATA AAC CTA GAA GAA GAG GAC GAT GAC AAC 48 Ala Ala Thr Ala Leu Glu He Asn Leu Glu Glu Glu Asp Asp Asp Asn 1 5 10 15
GAA GAC GAA GTA GAC GAG CAA GCT GAG CAG CAA AAA ACT CAC GTA TTT 96 Glu Asp Glu Val Asp Glu Gin Ala Glu Gin Gin Lys Thr His Val Phe 20 25 30
GGG CAG GCG CCT TAT TCT GGT ATA AAT ATT ACA AAG GAG 135
Gly Gin Ala Pro Tyr Ser Gly He Asn He Thr Lys Glu 35 40 45 (2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
Ala Ala Thr Ala Leu Glu He Asn Leu Glu Glu Glu Asp Asp Asp Asn 1 5 10 15
Glu Asp Glu Val Asp Glu Gin Ala Glu Gin Gin Lys Thr His Val Phe 20 25 30
Gly Gin Ala Pro Tyr Ser Gly He Asn He Thr Lys Glu 35 40 45
(2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:
TCA GAC AAT GCA GAA ACA CAA GCT AAA CCT GTA 33
Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val 1 5 10
(2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 11 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val 1 5 10
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15: GTC GAA GGT CAA ACA CCT AAA 21
Val Glu Gly Gin Thr Pro Lys 1 5
(2) INFORMATION FOR SEQ ID NO: 16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:
Val Glu Gly Gin Thr Pro Lys 1 5
(2) INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
AAC GAA GCT GAT GCT AAT GCG GCA 24
Asn Glu Ala Asp Ala Asn Ala Ala 1 5
(2) INFORMATION FOR SEQ ID NO: 18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:
Asn Glu Ala Asp Ala Asn Ala Ala 1 5
(2) INFORMATION FOR SEQ ID NO: 19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:
TAC GAA ACT GAA ATT AAT CAT GCA 24
Tyr Glu Thr Glu He Asn His Ala 1 5 (2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20:
Tyr Glu Thr Glu He Asn His Ala 1 5
(2) INFORMATION FOR SEQ ID NO: 21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21:
TCC GTT CTG GTT CCG GAT GAA AAA GGG GTG CCT CTT CCA AAG 42
Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys 1 5 10
(2) INFORMATION FOR SEQ ID NO: 22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:
Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys 1 5 10
(2) INFORMATION FOR SEQ ID NO: 23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23:
GGC ATT CTT GTA AAG CAA CAA AAT GGA AAG CTA GAA AGT CAA 42
Gly He Leu Val Lys Gin Gin Asn Gly Lys Leu Glu Ser Gin 1 5 10
(2) INFORMATION FOR SEQ ID NO: 24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:
Gly He Leu Val Lys Gin Gin Asn Gly Lys Leu Glu Ser Gin 1 5 10
(2) INFORMATION FOR SEQ ID NO:25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 51 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:
TCA AAT ACT ACC TCT TTG AAC GAC CGG CAA GGC AAT GCT ACT AAA CCA 48 Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin Gly Asn Ala Thr Lys Pro 1 5 10 15
AAA 51
Lys
(2) INFORMATION FOR SEQ ID NO: 26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 17 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26:
Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin Gly Asn Ala Thr Lys Pro 1 5 10 15
Lys
(2) INFORMATION FOR SEQ ID NO: 27:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 48 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:27:
TCA ACT ACT GAG GCG ACC GCA GGC AAT GGT GAT AAC TTG ACT CCT AAA 48 Ser Thr Thr Glu Ala Thr Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys 1 5 10 15
(2) INFORMATION FOR SEQ ID NO: 28:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 16 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28:
Ser Thr Thr Glu Ala Thr Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys 1 5 10 15
(2) INFORMATION FOR SEQ ID NO: 29:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:29:
TTG TAC AGT GAA GAT GTA AAT ATG 24
Leu Tyr Ser Glu Asp Val Asn Met
1 5
(2) INFORMATION FOR SEQ ID NO: 30:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30:
Leu Tyr Ser Glu Asp Val Asn Met 1 5
(2) INFORMATION FOR SEQ ID NO: 31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31:
TTG TAC AGT GAA GAT GTA GAT ATA 24
Leu Tyr Ser Glu Asp Val Asp He 1 5
(2) INFORMATION FOR SEQ ID NO: 32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 32:
Leu Tyr Ser Glu Asp Val Asp He 1 5
(2) INFORMATION FOR SEQ ID NO: 33:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:33:
GGA AAA GGT GAT GAA AAT TCT AAA GCT ATG TTG GGT 36
Gly Lys Gly Asp Glu Asn Ser Lys Ala Met Leu Gly 1 5 10
(2) INFORMATION FOR SEQ ID NO: 34:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 34:
Gly Lys Gly Asp Glu Asn Ser Lys Ala Met Leu Gly 1 5 10
(2) INFORMATION FOR SEQ ID NO: 35:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 35:
ACT ATT AAG GAA GGT AAC TCA CGA GAA CTA ATG GGC 36
Thr He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly 1 5 10
(2) INFORMATION FOR SEQ ID NO: 36:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:36: Thr He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly 1 5 10
(2) INFORMATION FOR SEQ ID NO: 37:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 165 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 37:
AAT TAT TGT TTT CCT CTT GGG GGT ATT GGG GTA ACT GAC ACC TAT CAA 48 Asn Tyr Cys Phe Pro Leu Gly Gly He Gly Val Thr Asp Thr Tyr Gin 1 5 10 15
GCT ATT AAG GCT AAT GGC AAT GGC TCA GGC GAT AAT GGA GAT ACT ACA 96 Ala He Lys Ala Asn Gly Asn Gly Ser Gly Asp Asn Gly Asp Thr Thr 20 25 30
TGG ACA AAA GAT GAA ACT TTT GCA ACA CGT AAT GAA ATA GGA GTG GGT 144 Trp Thr Lys Asp Glu Thr Phe Ala Thr Arg Asn Glu He Gly Val Gly 35 40 45
AAC AAC TTT GCC ATG GAA ATT 165
Asn Asn Phe Ala Met Glu He 50 55
(2) INFORMATION FOR SEQ ID NO: 38:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 55 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:38:
Asn Tyr Cys Phe Pro Leu Gly Gly He Gly Val Thr Asp Thr Tyr Gin 1 5 10 15
Ala He Lys Ala Asn Gly Asn Gly Ser Gly Asp Asn Gly Asp Thr Thr 20 25 30
Trp Thr Lys Asp Glu Thr Phe Ala Thr Arg Asn Glu He Gly Val Gly 35 40 45
Asn Asn Phe Ala Met Glu He 50 55
(2) INFORMATION FOR SEQ ID NO:39:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 153 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 39:
AAT TAC TGC TTT CCA CTG GGA GGT GTG ATT AAT ACA GAG ACT CTT ACC 48 Asn Tyr Cys Phe Pro Leu Gly Gly Val He Asn Thr Glu Thr Leu Thr 1 5 10 15
AAG GTA AAA CCT AAA ACA GGT CAG GAA AAT GGA TGG GAA AAA GAT GCT 96 Lys Val Lys Pro Lys Thr Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala 20 25 30
ACA GAA TTT TCA GAT AAA AAT GAA ATA AGA GTT GGA AAT AAT TTT GCC 144 Thr Glu Phe Ser Asp Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala 35 40 45
ATG GAA ATC 153
Met Glu He 50
(2) INFORMATION FOR SEQ ID NO: 40:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 51 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 40:
Asn Tyr Cys Phe Pro Leu Gly Gly Val He Asn Thr Glu Thr Leu Thr 1 5 10 15
Lys Val Lys Pro Lys Thr Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala 20 25 30
Thr Glu Phe Ser Asp Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala 35 40 45
Met Glu He 50
(2) INFORMATION FOR SEQ ID NO: 41:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 54 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 41 :
GTA ACT GAC ACC TAT CAA GCT ATT AAG GCT AAT GGC AAT GGC TCA GGC 48 Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser Gly 1 5 10 15
GAT AAT 54
Asp Asn
(2) INFORMATION FOR SEQ ID NO: 42:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42:
Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser Gly 1 5 10 15
Asp Asn
(2) INFORMATION FOR SEQ ID NO: 43:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 87 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 43:
AAT ACA GAG ACT CTT ACC AAG GTA AAA CCT AAA ACA GGT CAG GAA AAT 48 Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys Thr Gly Gin Glu Asn 1 5 10 15
GGA TGG GAA AAA GAT GCT ACA GAA TTT TCA GAT AAA AAT 87
Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp Lys Asn 20 25
(2) INFORMATION FOR SEQ ID NO : 44 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 44:
Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys Thr Gly Gin Glu Asn
1 5 10 15 -
Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp Lys Asn 20 25
(2) INFORMATION FOR SEQ ID NO: 45:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 45:
ACT TTT GCA ACA CGT AAT GAA 21
Thr Phe Ala Thr Arg Asn Glu 1 5 (2) INFORMATION FOR SEQ ID NO : 46 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:46:
Thr Phe Ala Thr Arg Asn Glu 1 5
(2) INFORMATION FOR SEQ ID NO: 47:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 47:
ACA GAA TTT TCA GAT AAA AAT GAA 24
Thr Glu Phe Ser Asp Lys Asn Glu 1 5
(2) INFORMATION FOR SEQ ID NO: 48:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 48:
Thr Glu Phe Ser Asp Lys Asn Glu 1 5
(2) INFORMATION FOR SEQ ID NO: 49:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
GAC TAC AAA GAC GAC GAC GAC AAA 24
Asp Tyr Lys Asp Asp Asp Asp Lys 1 5
(2) INFORMATION FOR SEQ ID NO: 50:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 8 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 50:
Asp Tyr Lys Asp Asp Asp Asp Lys 1 5
(2) INFORMATION FOR SEQ ID NO: 51:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2907 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 51:
ATG GCT ACC CCT TCG ATG ATG CCG CAG TGG TCT TAC ATG CAC ATC TCG 48 Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser 1 5 10 15
GGC CAG GAC GCC TCG GAG TAC CTG AGC CCC GGG CTG GTG CAG TTT GCC 96 Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
CGC GCC ACC GAG ACG TAC TTC AGC CTG AAT AAC AAG TTT AGA AAC CCC 144 Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
ACG GTG GCA CCT ACG CAC GAC GTA ACC ACA GAC CGG TCC CAG CGT TTG 192 Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
ACG CTG CGG TTC ATC CCT GTG GAC CGC GAG GAT ACC GCG TAC TCG TAC 240 Thr Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75
AAA GCG CGG TTC ACC CTG GCT GTG GGT GAC AAC CGT GTG CTT GAT ATG 288 Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 80 85 90 95
GCT TCC ACG TAC TTT GAC ATC CGC GGC GTG CTG GAC AGG GGG CCT ACT 336 Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
TTT AAG CCC TAC TCC GGC ACT GCC TAC AAC GCT CTA GCT CCC AAG GGC 384 Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
GCT CCT AAC TCC TGT GAG TGG GAA CAA ACC GAA GAT AGC GGC CGG GCA 432 Ala Pro Asn Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala 130 135 140
GTT GCC GAG GAT GAA GAA GAG GAA GAT GAA GAT GAA GAA GAG GAA GAA 480 Val Ala Glu Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu 145 150 155 GAA GAG CAA AAC GCT CGA GAT CAG GCT ACT AAG AAA ACA CAT GTC TAT 528 Glu Glu Gin Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr 160 165 170 175
GCC CAG GCT CCT TTG TCT GGA GAA ACA ATT ACA AAA AGC GGG CTA CAA 576 Ala Gin Ala Pro Leu Ser Gly Glu Thr He Thr Lys Ser Gly Leu Gin 180 185 190
ATA GGA TCA GAC AAT GCA GAA ACA CAA GCT AAA CCT GTA TAC GCA GAT 624 He Gly Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp 195 200 205
CCT TCC TAT CAA CCA GAA CCT CAA ATT GGC GAA TCT CAG TGG AAC GAA 672 Pro Ser Tyr Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp Asn Glu 210 215 220
GCT GAT GCT AAT GCG GCA GGA GGG AGA GTG CTT AAA AAA ACA ACT CCC 720 Ala Asp Ala Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro 225 230 235
ATG AAA CCA TGC TAT GGA TCT TAT GCC AGG CCT ACA AAT CCT TTT GGT 768 Met Lys Pro Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly 240 245 250 255
GGT CAA TCC GTT CTG GTT CCG GAT GAA AAA GGG GTG CCT CTT CCA AAG 816 Gly Gin Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys 260 265 270
GTT GAC TTG CAA TTC TTC TCA AAT ACT ACC TCT TTG AAC GAC CGG CAA 864 Val Asp Leu Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin 275 280 285
GGC AAT GCT ACT AAA CCA AAA GTG GTT TTG TAC AGT GAA GAT GTA AAT 912 Gly Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn 290 295 300
ATG GAA ACC CCA GAC ACA CAT CTG TCT TAC AAA CCT GGA AAA GGT GAT 960 Met Glu Thr Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp 305 310 315
GAA AAT TCT AAA GCT ATG TTG GGT CAA CAA TCT ATG CCA AAC AGA CCC 1008 Glu Asn Ser Lys Ala Met Leu Gly Gin Gin Ser Met Pro Asn Arg Pro 320 325 330 335
AAT TAC ATT GCT TTC AGG GAC AAT TTT ATT GGC CTA ATG TAT TAT AAC 1056 Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu Met Tyr Tyr Asn 340 345 350
AGC ACT GGC AAC ATG GGT GTT CTT GCT GGT CAG GCA TCG CAG CTA AAT 1104 Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala Ser Gin Leu Asn 355 360 365
GCC GTG GTA GAT TTG CAA GAC AGA AAC ACA GAG CTG TCC TAT CAA CTC 1152 Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu Ser Tyr Gin Leu 370 375 380
TTG CTT GAT TCC ATA GGT GAT AGA ACC AGA TAT TTT TCT ATG TGG AAT 1200 Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe Ser Met Trp Asn 385 390 395
CAG GCT GTA GAC AGC TAT GAT CCA GAT GTT AGA ATC ATT GAA AAC CAT 1248 Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He He Glu Asn His 400 405 410 415 GGA ACT GAG GAT GAA TTG CCA AAT TAT TGT TTT CCT CTT GGG GGT ATT 1296 Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro Leu Gly Gly He 420 425 430
GGG GTA ACT GAC ACC TAT CAA GCT ATT AAG GCT AAT GGC AAT GGC TCA 1344 Gly Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser 435 440 445
GGC GAT AAT GGA GAT ACT ACA TGG ACA AAA GAT GAA ACT TTT GCA ACA 1392 Gly Asp Asn Gly Asp Thr Thr Trp Thr Lys Asp Glu Thr Phe Ala Thr 450 455 460
CGT AAT GAA ATA GGA GTG GGT AAC AAC TTT GCC ATG GAA ATT AAC CTA 1440 Arg Asn Glu He Gly Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 465 470 475
AAT GCC AAC CTA TGG AGA AAT TTC CTT TAC TCC AAT ATT GCG CTG TAC 1488 Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 480 485 490 495
CTG CCA GAC AAG CTA AAA TAC AAC CCC ACC AAT GTG GAA ATA TCT GAC 1536 Leu Pro Asp Lys Leu Lys Tyr Asn Pro Thr Asn Val Glu He Ser Asp 500 505 510
AAC CCC AAC ACC TAC GAC TAC ATG AAC AAG CGA GTG GTG GCT CCC GGG 1584 Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 515 520 525
CTT GTA GAC TGC TAC ATT AAC CTT GGG GCG CGC TGG TCT CTG GAC TAC 1632 Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 530 535 540
ATG GAC AAC GTT AAT CCC TTT AAC CAC CAC CGC AAT GCG GGC CTC CGT 1680 Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 545 550 555
TAT CGC TCC ATG TTG TTG GGA AAC GGC CGC TAC GTG CCC TTT CAC ATT 1728 Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 560 565 570 575
CAG GTG CCC CAA AAG TTT TTT GCC ATT AAA AAC CTC CTC CTC CTG CCA 1776 Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 580 585 590
GGC TCA TAT ACA TAT GAA TGG AAC TTC AGG AAG GAT GTT AAC ATG GTT 1824 Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 595 600 605
CTG CAG AGC TCT CTG GGA AAC GAT CTT AGA GTT GAC GGG GCT AGC ATT 1872 Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 610 615 620
AAG TTT GAC AGC ATT TGT CTT TAC GCC ACC TTC TTC CCC ATG GCC CAC 1920 Lys Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His 625 630 635
AAC ACG GCC TCC ACG CTG GAA GCC ATG CTC AGA AAT GAC ACC AAC GAC 1968 Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 640 645 650 655
CAG TCC TTT AAT GAC TAC CTT TCC GCC GCC AAC ATG CTA TAC CCC ATA 2016 Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 660 665 670 CCC GCC AAC GCC ACC AAC GTG CCC ATC TCC ATC CCA TCG CGC AAC TGG 2064 Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 675 680 685
GCA GCA TTT CGC GGT TGG GCC TTC ACA CGC TTG AAG ACA AAG GAA ACC 2112 Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 690 695 700
CCT TCC CTG GGA TCA GGC TAC GAC CCT TAC TAC ACC TAC TCT GGC TCC 2160 Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 705 710 715
ATA CCA TAC CTT GAC GGA ACC TTC TAT CTT AAT CAC ACC TTT AAG AAG 2208 He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 720 725 730 735
GTG GCC ATT ACC TTT GAC TCT TCT GTT AGC TGG CCG GGC AAC GAC CGC 2256 Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 740 745 750
CTG CTT ACT CCC AAT GAG TTT GAG ATT AAA CGC TCA GTT GAC GGG GAG 2304 Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 755 760 765
GGC TAC AAC GTA GCT CAG TGC AAC ATG ACC AAG GAC TGG TTC CTG GTG 2352 Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 770 775 780
CAG ATG TTG GCC AAC TAC AAT ATT GGC TAC CAG GGC TTC TAC ATT CCA 2400 Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 785 790 795
GAA AGC TAC AAG GAC CGC ATG TAC TCG TTC TTC AGA AAC TTC CAG CCC 2448 Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 800 805 810 815
ATG AGC CGG CAA GTG GTT GAC GAT ACT AAA TAC AAG GAG TAT CAG CAG 2496 Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Glu Tyr Gin Gin 820 825 830
GTT GGA ATT CTT CAC CAG CAT AAC AAC TCA GGA TTC GTA GGC TAC CTC 2544 Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 835 840 845
GCT CCC ACC ATG CGC GAG GGA CAG GCT TAC CCC GCC AAC GTG CCC TAC 2592 Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Val Pro Tyr 850 855 860
CCA CTA ATA GGC AAA ACC GCG GTT GAC AGT ATT ACC CAG AAA AAG TTT 2640 Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 865 870 875
CTT TGC GAT CGC ACC CTT TGG CGC ATC CCA TTC TCC AGT AAC TTT ATG 2688 Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 880 885 890 895
TCC ATG GGC GCA CTC ACA GAC CTG GGC CAA AAC CTT CTC TAC GCC AAC 2736 Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 900 905 910
TCC GCC CAC GCG CTA GAC ATG ACT TTT GAG GTG GAT CCC ATG GAC GAG 2784 Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 915 920 925 CCC ACC CTT CTT TAT GTT TTG TTT GAA GTC TTT GAC GTG GTC CGT GTG 2832 Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 930 935 940
CAC CAG CCG CAC CGC GGC GTC ATC GAG ACC GTG TAC CTG CGC ACG CCC 2880 His Gin Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 945 950 955
TTC TCG GCC GGC AAC GCC ACA ACA TAA 2907
Phe Ser Ala Gly Asn Ala Thr Thr 960 965
(2) INFORMATION FOR SEQ ID NO: 52:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 967 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 52:
Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser Gly 1 5 10 15
Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala Arg 20 25 30
Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro Thr 35 40 45
Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu Thr 50 55 60
Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr Lys 65 70 75 80
Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met Ala 85 90 95
Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr Phe 100 105 110
Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly Ala 115 120 125
Pro Asn Ser Cys Glu Trp Glu Gin Thr Glu Asp Ser Gly Arg Ala Val 130 135 140
Ala Glu Asp Glu Glu Glu Glu Asp Glu Asp Glu Glu Glu Glu Glu Glu 145 150 155 160
Glu Gin Asn Ala Arg Asp Gin Ala Thr Lys Lys Thr His Val Tyr Ala 165 170 175
Gin Ala Pro Leu Ser Gly Glu Thr He Thr Lys Ser Gly Leu Gin He 180 185 190
Gly Ser Asp Asn Ala Glu Thr Gin Ala Lys Pro Val Tyr Ala Asp Pro 195 200 205
Ser Tyr Gin Pro Glu Pro Gin He Gly Glu Ser Gin Trp Asn Glu Ala 210 215 220 Asp Ala Asn Ala Ala Gly Gly Arg Val Leu Lys Lys Thr Thr Pro Met 225 230 235 240
Lys Pro Cys Tyr Gly Ser Tyr Ala Arg Pro Thr Asn Pro Phe Gly Gly 245 250 255
Gin Ser Val Leu Val Pro Asp Glu Lys Gly Val Pro Leu Pro Lys Val 260 265 270
Asp Leu Gin Phe Phe Ser Asn Thr Thr Ser Leu Asn Asp Arg Gin Gly 275 280 285
Asn Ala Thr Lys Pro Lys Val Val Leu Tyr Ser Glu Asp Val Asn Met 290 295 300
Glu Thr Pro Asp Thr His Leu Ser Tyr Lys Pro Gly Lys Gly Asp Glu 305 310 315 320
Asn Ser Lys Ala Met Leu Gly Gin Gin Ser Met Pro Asn Arg Pro Asn 325 330 335
Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu Met Tyr Tyr Asn Ser 340 345 350
Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala Ser Gin Leu Asn Ala 355 360 365
Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu Ser Tyr Gin Leu Leu 370 375 380
Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe Ser Met Trp Asn Gin 385 390 395 400
Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He He Glu Asn His Gly 405 410 415
Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro Leu Gly Gly He Gly 420 425 430
Val Thr Asp Thr Tyr Gin Ala He Lys Ala Asn Gly Asn Gly Ser Gly 435 440 445
Asp Asn Gly Asp Thr Thr Trp Thr Lys Asp Glu Thr Phe Ala Thr Arg 450 455 460
Asn Glu He Gly Val Gly Asn Asn Phe Ala Met Glu He Asn Leu Asn 465 470 475 480
Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr Leu 485 490 495
Pro Asp Lys Leu Lys Tyr Asn Pro Thr Asn Val Glu He Ser Asp Asn 500 505 510
Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly Leu 515 520 525
Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr Met 530 535 540
Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg Tyr 545 550 555 560 Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He Gin 565 570 575
Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro Gly 580 585 590
Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val Leu 595 600 605
Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He Lys 610 615 620
Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His Asn 625 630 635 640
Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp Gin 645 650 655
Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He Pro 660 665 670
Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp Ala 675 680 685
Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr Pro 690 695 700
Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser He 705 710 715 720
Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys Val 725 730 735
Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg Leu 740 745 750
Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu Gly 755 760 765
Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val Gin 770 775 780
Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro Glu 785 790 795 800
Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro Met 805 810 815
Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Glu Tyr Gin Gin Val 820 825 830
Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu Ala 835 840 845
Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Val Pro Tyr Pro 850 855 860
Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe Leu 865 870 875 880
Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met Ser 885 890 895 Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn Ser 900 905 910
Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu Pro 915 920 925
Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val His 930 935 940
Gin Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro Phe 945 950 955 960
Ser Ala Gly Asn Ala Thr Thr 965
(2) INFORMATION FOR SEQ ID NO: 53:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2858 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 53:
ATG GCT ACC CCT TCG ATG ATG CCG CAG TGG TCT TAC ATG CAC ATC TCG 48 Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser 1 5 10 15
GGC CAG GAC GCC TCG GAG TAC CTG AGC CCC GGG CTG GTG CAG TTT GCC 96 Gly Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala 20 25 30
CGC GCC ACC GAG ACG TAC TTC AGC CTG AAT AAC AAG TTT AGA AAC CCC 144 Arg Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro 35 40 45
ACG GTG GCG CCT ACG CAC GAC GTG ACC ACA GAC CGG TCC CAG CGT TTG 192 Thr Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu 50 55 60
ACG CTG CGG TTC ATC CCT GTG GAC CGT GAG GAT ACT GCG TAC TCG TAC 240 Thr Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr 65 70 75
AAG GCG CGG TTC ACC CTA GCT GTG GGT GAT AAC CGT GTG CTG GAC ATG 288 Lys Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met 80 85 90 95
GCT TCC ACG TAC TTT GAC ATC CGC GGC GTG CTG GAC AGG GGC CCT ACT 336 Ala Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr 100 105 110
TTT AAG CCC TAC TCT GGC ACT GCC TAC AAC GCC CTG GCT CCC AAG GGT 384 Phe Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly 115 120 125
GCC CCA AAT CCT TGC GAA TGG GAT GAA GCT GCT ACT GCT CTT GAA ATA 432 Ala Pro Asn Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He 130 135 140
AAC CTA GAA GAA GAG GAC GAT GAC AAC GAA GAC GAA GTA GAC GAG CAA 480 Asn Leu Glu Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin 145 150 155
GCT GAG CAG CAA AAA ACT CAC GTA TTT GGG CAG GCG CCT TAT TCT GGT 528 Ala Glu Gin Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly 160 165 170 175
ATA AAT ATT ACA AAG GAG GGT ATT CAA ATA GGT GTC GAA GGT CAA ACA 576 He Asn He Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr 180 185 190
CCT AAA TAT GCC GAT AAA ACA TTT CAA CCT GAA CCT CAA ATA GGA GAA 624 Pro Lys Tyr Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu 195 200 205
TCT CAG TGG TAC GAA ACT GAA ATT AAT CAT GCA GCT GGG AGA GTC CTT 672 Ser Gin Trp Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu 210 215 220
AAA AAG ACT ACC CCA ATG AAA CCA TGT TAC GGT TCA TAT GCA AAA CCC 720 Lys Lys Thr Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro 225 230 235
ACA AAT GAA AAT GGA GGG CAA GGC ATT CTT GTA AAG CAA CAA AAT GGA 768 Thr Asn Glu Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly 240 245 250 255
AAG CTA GAA AGT CAA GTG GAA ATG CAA TTT TTC TCA ACT ACT GAG GCG 816 Lys Leu Glu Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala 260 265 270
ACC GCA GGC AAT GGT GAT AAC TTG ACT CCT AAA GTG GTA TTG TAC AGT 864 Thr Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser 275 280 285
GAA GAT GTA GAT ATA GAA ACC CCA GAC ACT CAT ATT TCT TAC ATG CCC 912 Glu Asp Val Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro 290 295 300
ACT ATT AAG GAA GGT AAC TCA CGA GAA CTA ATG GGC CAA CAA TCT ATG 960 Thr He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met 305 310 315
CCC AAC AGG CCT AAT TAC ATT GCT TTT AGG GAC AAT TTT ATT GGT CTA 1008 Pro Asn Arg Pro Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu 320 325 330 335
ATG TAT TAC AAC AGC ACG GGT AAT ATG GGT GTT CTG GCG GGC CAA GCA 1056 Met Tyr Tyr Asn Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala 340 345 350
TCG CAG TTG AAT GCT GTT GTA GAT TTG CAA GAC AGA AAC ACA GAG CTT 1104 Ser Gin Leu Asn Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu 355 360 365
TCA TAC CAG CTT TTG CTT GAT TCC ATT GGT GAT AGA ACC AGG TAC TTT 1152 Ser Tyr Gin Leu Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe 370 375 380
TCT ATG TGG AAT CAG GCT GTT GAC AGC TAT GAT CCA GAT GTT AGA ATT 1200 Ser Met Trp Asn Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He 385 390 395
ATT GAA AAT CAT GGA ACT GAA GAT GAA CTT CCA AAT TAC TGC TTT CCA 1248 He Glu Asn His Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro 400 405 410 415
CTG GGA GGT GTG ATT AAT ACA GAG ACT CTT ACC AAG GTA AAA CCT AAA 1296 Leu Gly Gly Val He Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys 420 425 430
ACA GGT CAG GAA AAT GGA TGG GAA AAA GAT GCT ACA GAA TTT TCA GAT 1344 Thr Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp 435 440 445
AAA AAT GAA ATA AGA GTT GGA AAT AAT TTT GCC ATG GAA ATC AAT CTA 1392 Lys Asn Glu He Arg Val Gly Asn Asn Phe Ala Met Glu He Asn Leu 450 455 460
AAT GCC AAC CTG TGG AGA AAT TTC CTG TAC TCC AAC ATA GCG CTG TAT 1440 Asn Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr 465 470 475
TTG CCC GAC AAG CTA AAG TAC AGT CCT TCC AAC GTA AAA ATT TCT GAT 1488 Leu Pro Asp Lys Leu Lys Tyr Ser Pro Ser Asn Val Lys He Ser Asp 480 485 490 495
AAC CCA AAC ACC TAC GAC TAC ATG AAC AAG CGA GTG GTG GCT CCC GGG 1536 Asn Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly 500 505 510
TTA GTG GAC TGC TAC ATT AAC CTT GGA GCA CGC TGG TCC CTT GAC TAT 1584 Leu Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr 515 520 525
ATG GAC AAC GTC AAC CCA TTT AAC CAC CAC CGC AAT GCT GGC CTG CGC 1632 Met Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg 530 535 540
TAC CGC TCA ATG TTG CTG GGC AAT GGT CGC TAT GTG CCC TTC CAC ATC 1680 Tyr Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He 545 550 555
CAG GTG CCT CAG AAG TTC TTT GCC ATT AAA AAC CTC CTT CTC CTG CCG 1728 Gin Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro 560 565 570 575
GGC TCA TAC ACC TAC GAG TGG AAC TTC AGG AAG GAT GTT AAC ATG GTT 1776 Gly Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val 580 585 590
CTG CAG AGC TCC CTA GGA AAT GAC CTA AGG GTT GAC GGA GCC AGC ATT 1824 Leu Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He 595 600 605
AAG TTT GAT AGC ATT TGC CTT TAC GCC ACC TTC TTC CCC ATG GCC CAC 1872 Lys Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His 610 615 620
AAC ACC GCC TCC ACG CTT GAG GCC ATG CTT AGA AAC GAC ACC AAC GAC 1920 Asn Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp 625 630 635
CAG TCC TTT AAC GAC TAT CTC TCC GCC GCC AAC ATG CTC TAC CCT ATA 1968 Gin Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He 640 645 650 655 CCC GCC AAC GCT ACC AAC GTG CCC ATA TCC ATC CCC TCC CGC AAC TGG 2016 Pro Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp 660 665 670
GCG GCT TTC CGC GGC TGG GCC TTC ACG CGC CTT AAG ACT AAG GAA ACC 2064 Ala Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr 675 680 685
CCA TCA CTG GGC TCG GGC TAC GAC CCT TAT TAC ACC TAC TCT GGC TCT 2112 Pro Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser 690 695 700
ATA CCC TAC CTA GAT GGA ACC TTT TAC CTC AAC CAC ACC TTT AAG AAG 2160 He Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys 705 710 715
GTG GCC ATT ACC TTT GAC TCT TCT GTC AGC TGG CCT GGC AAT GAC CGC 2208 Val Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg 720 725 730 735
CTG CTT ACC CCC AAC GAG TTT GAA ATT AAG CGC TCA GTT GAC GGG GAG 2256 Leu Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu 740 745 750
GGT TAC AAC GTT GCC CAG TGT AAC ATG ACC AAA GAC TGG TTC CTG GTA 2304 Gly Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val 755 760 765
CAA ATG CTA GCT AAC TAC AAC ATT GGC TAC CAG GGC TTC TAT ATC CCA 2352 Gin Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro 770 775 780
GAG AGC TAC AAG GAC CGC ATG TAC TCC TTC TTT AGA AAC TTC CAG CCC 2400 Glu Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro 785 790 795
ATG AGC CGT CAG GTG GTG GAT GAT ACT AAA TAC AAG GAC TAC CAA CAG 2448 Met Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Asp Tyr Gin Gin 800 805 810 815
GTG GGC ATC CTA CAC CAA CAC AAC AAC TCT GGA TTT GTT GGC TAC CTT 2496 Val Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu 820 825 830
GCC CCC ACC ATG CGC GAA GGA CAG GCC TAC CCT GCT AAC TTC CCC TAT 2544 Ala Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Phe Pro Tyr 835 840 845
CCG CTT ATA GGC AAG ACC GCA GTT GAC AGC ATT ACC CAG AAA AAG TTT 2592 Pro Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe 850 855 860
CTT TGC GAT CGC ACC CTT TGG CGC ATC CCA TTC TCC AGT AAC TTT ATG 2640 Leu Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met 865 870 875
TCC ATG GGC GCA CTC ACA GAC CTG GGC CAA AAC CTT CTC TAC GCC AAC 2688 Ser Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn 880 885 890 895
TCC GCC CAC GCG CTA GAC ATG ACT TTT GAG GTG GAT CCC ATG GAC GAG 2736 Ser Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu 900 905 910 CCC ACC CTT CTT TAT GTT TTG TTT GAA GTC TTT GAC GTG GTC CGT GTG 2784 Pro Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val 915 920 925
CAC CGG CCG CAC CGC GGC GTC ATC GAA ACC GTG TAC CTG CGC ACG CCC 2832 His Arg Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro 930 935 940
TTC TCG GCC GGC AAC GCA CAA CAT AA 2858
Phe Ser Ala Gly Asn Ala Gin His 945 950
(2) INFORMATION FOR SEQ ID NO: 54:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 951 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 54:
Ala Thr Pro Ser Met Met Pro Gin Trp Ser Tyr Met His He Ser Gly 1 5 10 15
Gin Asp Ala Ser Glu Tyr Leu Ser Pro Gly Leu Val Gin Phe Ala Arg 20 25 30
Ala Thr Glu Thr Tyr Phe Ser Leu Asn Asn Lys Phe Arg Asn Pro Thr 35 40 45
Val Ala Pro Thr His Asp Val Thr Thr Asp Arg Ser Gin Arg Leu Thr 50 55 60
Leu Arg Phe He Pro Val Asp Arg Glu Asp Thr Ala Tyr Ser Tyr Lys 65 70 75 80
Ala Arg Phe Thr Leu Ala Val Gly Asp Asn Arg Val Leu Asp Met Ala 85 90 95
Ser Thr Tyr Phe Asp He Arg Gly Val Leu Asp Arg Gly Pro Thr Phe 100 105 110
Lys Pro Tyr Ser Gly Thr Ala Tyr Asn Ala Leu Ala Pro Lys Gly Ala 115 120 125
Pro Asn Pro Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu He Asn 130 135 140
Leu Glu Glu Glu Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gin Ala 145 150 155 160
Glu Gin Gin Lys Thr His Val Phe Gly Gin Ala Pro Tyr Ser Gly He 165 170 175
Asn He Thr Lys Glu Gly He Gin He Gly Val Glu Gly Gin Thr Pro 180 185 190
Lys Tyr Ala Asp Lys Thr Phe Gin Pro Glu Pro Gin He Gly Glu Ser 195 200 205
Gin Trp Tyr Glu Thr Glu He Asn His Ala Ala Gly Arg Val Leu Lys 210 215 220 Lys Thr Thr Pro Met Lys Pro Cys Tyr Gly Ser Tyr Ala Lys Pro Thr 225 230 235 240
Asn Glu Asn Gly Gly Gin Gly He Leu Val Lys Gin Gin Asn Gly Lys 245 250 255
Leu Glu Ser Gin Val Glu Met Gin Phe Phe Ser Thr Thr Glu Ala Thr 260 265 270
Ala Gly Asn Gly Asp Asn Leu Thr Pro Lys Val Val Leu Tyr Ser Glu 275 280 285
Asp Val Asp He Glu Thr Pro Asp Thr His He Ser Tyr Met Pro Thr 290 295 300
He Lys Glu Gly Asn Ser Arg Glu Leu Met Gly Gin Gin Ser Met Pro 305 310 315 320
Asn Arg Pro Asn Tyr He Ala Phe Arg Asp Asn Phe He Gly Leu Met 325 330 335
Tyr Tyr Asn Ser Thr Gly Asn Met Gly Val Leu Ala Gly Gin Ala Ser 340 345 350
Gin Leu Asn Ala Val Val Asp Leu Gin Asp Arg Asn Thr Glu Leu Ser 355 360 365
Tyr Gin Leu Leu Leu Asp Ser He Gly Asp Arg Thr Arg Tyr Phe Ser 370 375 380
Met Trp Asn Gin Ala Val Asp Ser Tyr Asp Pro Asp Val Arg He He 385 390 395 400
Glu Asn His Gly Thr Glu Asp Glu Leu Pro Asn Tyr Cys Phe Pro Leu 405 410 415
Gly Gly Val He Asn Thr Glu Thr Leu Thr Lys Val Lys Pro Lys Thr 420 425 430
Gly Gin Glu Asn Gly Trp Glu Lys Asp Ala Thr Glu Phe Ser Asp Lys 435 440 445
Asn Glu He Arg Val Gly Asn Asn Phe Ala Met Glu He Asn Leu Asn 450 455 460
Ala Asn Leu Trp Arg Asn Phe Leu Tyr Ser Asn He Ala Leu Tyr Leu 465 470 475 480
Pro Asp Lys Leu Lys Tyr Ser Pro Ser Asn Val Lys He Ser Asp Asn 485 490 495
Pro Asn Thr Tyr Asp Tyr Met Asn Lys Arg Val Val Ala Pro Gly Leu 500 505 510
Val Asp Cys Tyr He Asn Leu Gly Ala Arg Trp Ser Leu Asp Tyr Met 515 520 525
Asp Asn Val Asn Pro Phe Asn His His Arg Asn Ala Gly Leu Arg Tyr 530 535 540
Arg Ser Met Leu Leu Gly Asn Gly Arg Tyr Val Pro Phe His He Gin 545 550 555 560 Val Pro Gin Lys Phe Phe Ala He Lys Asn Leu Leu Leu Leu Pro Gly 565 570 575
Ser Tyr Thr Tyr Glu Trp Asn Phe Arg Lys Asp Val Asn Met Val Leu 580 585 590
Gin Ser Ser Leu Gly Asn Asp Leu Arg Val Asp Gly Ala Ser He Lys 595 600 605
Phe Asp Ser He Cys Leu Tyr Ala Thr Phe Phe Pro Met Ala His Asn 610 615 620
Thr Ala Ser Thr Leu Glu Ala Met Leu Arg Asn Asp Thr Asn Asp Gin 625 630 635 640
Ser Phe Asn Asp Tyr Leu Ser Ala Ala Asn Met Leu Tyr Pro He Pro 645 650 655
Ala Asn Ala Thr Asn Val Pro He Ser He Pro Ser Arg Asn Trp Ala 660 665 670
Ala Phe Arg Gly Trp Ala Phe Thr Arg Leu Lys Thr Lys Glu Thr Pro 675 680 685
Ser Leu Gly Ser Gly Tyr Asp Pro Tyr Tyr Thr Tyr Ser Gly Ser He 690 695 700
Pro Tyr Leu Asp Gly Thr Phe Tyr Leu Asn His Thr Phe Lys Lys Val 705 710 715 720
Ala He Thr Phe Asp Ser Ser Val Ser Trp Pro Gly Asn Asp Arg Leu 725 730 735
Leu Thr Pro Asn Glu Phe Glu He Lys Arg Ser Val Asp Gly Glu Gly 740 745 750
Tyr Asn Val Ala Gin Cys Asn Met Thr Lys Asp Trp Phe Leu Val Gin 755 760 765
Met Leu Ala Asn Tyr Asn He Gly Tyr Gin Gly Phe Tyr He Pro Glu 770 775 780
Ser Tyr Lys Asp Arg Met Tyr Ser Phe Phe Arg Asn Phe Gin Pro Met 785 790 795 800
Ser Arg Gin Val Val Asp Asp Thr Lys Tyr Lys Asp Tyr Gin Gin Val 805 810 815
Gly He Leu His Gin His Asn Asn Ser Gly Phe Val Gly Tyr Leu Ala 820 825 830
Pro Thr Met Arg Glu Gly Gin Ala Tyr Pro Ala Asn Phe Pro Tyr Pro 835 840 845
Leu He Gly Lys Thr Ala Val Asp Ser He Thr Gin Lys Lys Phe Leu 850 855 860
Cys Asp Arg Thr Leu Trp Arg He Pro Phe Ser Ser Asn Phe Met Ser 865 870 875 880
Met Gly Ala Leu Thr Asp Leu Gly Gin Asn Leu Leu Tyr Ala Asn Ser 885 890 895 Ala His Ala Leu Asp Met Thr Phe Glu Val Asp Pro Met Asp Glu Pro 900 905 910
Thr Leu Leu Tyr Val Leu Phe Glu Val Phe Asp Val Val Arg Val His 915 920 925
Arg Pro His Arg Gly Val He Glu Thr Val Tyr Leu Arg Thr Pro Phe 930 935 940
Ser Ala Gly Asn Ala Gin His 945 950
(2) INFORMATION FOR SEQ ID NO: 55:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 98 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 55:
GAA CTC GGA GGT GGA GGT GGA ACT AGT TTT GGA CGC GGA GAC ATT CGC 48 Glu Leu Gly Gly Gly Gly Gly Thr Ser Phe Gly Arg Gly Asp He Arg 1 5 10 15
AAT TAAAGTACTG GATTCATGAC TCTAGACTTA ATTAAGGATC CAATAAA 98
Asn
(2) INFORMATION FOR SEQ ID NO:56:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 17 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 56:
Glu Leu Gly Gly Gly Gly Gly Thr Ser Phe Gly Arg Gly Asp He Arg 1 5 10 15
Asn

Claims

WHAT IS CLAIMED IS:
1. A chimeric adenovirus coat protein comprising a nonnative amino acid sequence, wherein said chimeric adenovirus coat protein has a decreased ability or inability to be recognized by a neutralizing antibody directed against the wild-type adenovirus coat protein.
2. The chimeric adenovirus coat protein of claim 1, wherein said nonnative amino acid sequence comprises a deletion, insertion, or a replacement of a region of from about 1 to about 750 amino acids of said wild-type adenovirus coat protein.
3. The chimeric adenovirus coat protein of claim 1 or 2, wherein said nonnative amino acid sequence comprises a plurality of deletions, insertions, and/or replacements.
4. The chimeric adenovirus coat protein of any of claims 1-3, wherein said coat protein is a chimeric adenovirus hexon protein.
5. The chimeric adenovirus coat protein of claim 4, wherein said region deleted or replaced comprises a hypervariable region in either the II loop or the 12 loop.
6. The chimeric adenovirus coat protein of claim 5, wherein said hypervariable region is selected from the group consisting of HVRl, HVR2, HVR3, HVR4 , HVR5, HVR6, and HVR7.
7. The chimeric adenovirus coat protein of any of claims 1-6, comprising a sequence selected from the group consisting of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID N0:22, SEQ ID NO:24, SEQ ID N0:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, and SEQ ID NO:48.
8. The chimeric adenovirus coat protein of any of claims 1-7, wherein said nonnative amino acid sequence comprises a spacer of about 1 to about 750 amino acids.
9. The chimeric coat adenovirus coat protein of claim 8, wherein said spacer comprises the sequence of SEQ ID NO:50.
10. The chimeric adenovirus coat protein of any of claims 1-9, comprising an amino acid sequence of a coat protein of another serotype of adenovirus.
11. The chimeric adenovirus coat protein of claim 10, wherein said coat protein of another serotype is a hexon protein.
12. An isolated or purified nucleic acid that encodes the chimeric adenovirus coat protein of any of claims 1-11.
13. The isolated or purified nucleic acid of claim 12 comprising a sequence selected from the group consisting of SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO.ll, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, and SEQ ID NO:47.
14. The isolated or purified nucleic acid of claim 12 or 13 comprising SEQ ID NO: 9.
15. An adenoviral vector that comprises the chimeric adenovirus coat protein of any of claims 1-11.
16. A method of genetically modifying a cell which comprises contacting said cell with the adenoviral vector of claim 15.
17. A host cell that comprises the chimeric adenovirus coat protein of any of claims 1-11.
18. A method of constructing an adenoviral vector that has a decreased ability or inability to be recognized by a neutralizing antibody directed against wild-type adenovirus coat protein, which method comprises obtaining an adenoviral vector comprising a wild-type adenovirus coat protein and replacing said wild-type adenovirus coat protein with the chimeric adenovirus coat protein of any of claims 1-11.
PCT/US1998/005033 1997-03-13 1998-03-13 Chimeric adenoviral coat protein and methods of using same WO1998040509A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002283628A CA2283628A1 (en) 1997-03-13 1998-03-13 Chimeric adenoviral coat protein and methods of using same
JP53987998A JP2001517080A (en) 1997-03-13 1998-03-13 Chimeric adenovirus coat protein and method of using same
AU64652/98A AU745252B2 (en) 1997-03-13 1998-03-13 Chimeric adenoviral coat protein and methods of using same
EP98910401A EP0973927A1 (en) 1997-03-13 1998-03-13 Chimeric adenoviral coat protein and methods of using same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/816,346 1997-03-13
US08/816,346 US6127525A (en) 1995-02-21 1997-03-13 Chimeric adenoviral coat protein and methods of using same

Publications (1)

Publication Number Publication Date
WO1998040509A1 true WO1998040509A1 (en) 1998-09-17

Family

ID=25220343

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/005033 WO1998040509A1 (en) 1997-03-13 1998-03-13 Chimeric adenoviral coat protein and methods of using same

Country Status (6)

Country Link
US (2) US6127525A (en)
EP (1) EP0973927A1 (en)
JP (1) JP2001517080A (en)
AU (1) AU745252B2 (en)
CA (1) CA2283628A1 (en)
WO (1) WO1998040509A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000003029A2 (en) * 1998-07-08 2000-01-20 Introgene B.V. Chimaeric adenoviruses
WO2000012738A1 (en) * 1998-08-27 2000-03-09 Aventis Pharma S.A. Targeted adenovirus vectors for delivery of heterologous genes
US6057155A (en) * 1995-11-28 2000-05-02 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US6576456B2 (en) 1995-02-21 2003-06-10 Cornell Research Foundation, Inc. Chimeric adenovirus fiber protein
US6849446B2 (en) 2000-05-31 2005-02-01 University Of Saskatchewan Modified bovine adenovirus having altered tropism
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US6951755B2 (en) 1994-09-08 2005-10-04 Genvec, Inc. Vectors and methods for gene transfer
WO2006040330A2 (en) 2004-10-13 2006-04-20 Crucell Holland B.V. Improved adenoviral vectors and uses thereof
WO2007148148A1 (en) * 2006-06-19 2007-12-27 Institut Gustave Roussy Inhibition of the liver tropism of adenoviral vectors
WO2009058564A2 (en) 2007-11-01 2009-05-07 Maxygen, Inc. Immunosuppressive polypeptides and nucleic acids
EP2168603A1 (en) 2003-11-14 2010-03-31 GenVec, Inc. Therapeutic regimen for treating cancer
WO2011047316A1 (en) 2009-10-16 2011-04-21 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Nucleic acid sequences encoding expandable hiv mosaic proteins
WO2011057248A2 (en) 2009-11-09 2011-05-12 Genvec, Inc. Simian adenovirus and methods of use
WO2011057254A2 (en) 2009-11-09 2011-05-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Simian adenoviral vector-based vaccines
WO2012021730A2 (en) 2010-08-11 2012-02-16 Genvec, Inc. Respiratory syncytial virus (rsv) vaccine
WO2012083297A2 (en) 2010-12-17 2012-06-21 Genvec, Inc. Adenoviral vectors with modified hexon regions
WO2012088041A1 (en) 2010-12-20 2012-06-28 Genvec, Inc. Adenoviral vector-based dengue fever vaccine
EP2325298A3 (en) * 2008-03-04 2012-08-29 The Trustees of The University of Pennsylvania Simian adenoviruses SAdV-36, -42.1, -42.2, AND -44 and uses thereof
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
US8323663B2 (en) 2005-11-10 2012-12-04 Genvec, Inc. Adenoviral vector-based foot-and-mouth disease vaccine
EP2558481A1 (en) * 2010-04-14 2013-02-20 Mogam Biotechnology Research Institute Hexon isolated from simian adenovirus serotype 19, hypervariable region thereof and chimeric adenovirus using the same
EP2567967A2 (en) 2004-04-12 2013-03-13 GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Method of using adenoviral vectors to induce an immune response
WO2013181128A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Modified serotype 28 adenoviral vectors
US8765146B2 (en) 2005-08-31 2014-07-01 Genvec, Inc. Adenoviral vector-based malaria vaccines
EP2824185A1 (en) 2003-02-24 2015-01-14 GenVec, Inc. Materials and methods for treating disorders of the ear
US9228205B2 (en) 2000-11-15 2016-01-05 Crucell Holland B.V. Complementing cell lines
EP3284821A1 (en) 2010-03-23 2018-02-21 Intrexon Corporation Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2018064523A1 (en) 2016-09-30 2018-04-05 Genvec, Inc. Adenovectors for delivery of therapeutic genetic material into t cells
EP3269390A3 (en) * 2004-01-23 2018-04-18 MSD Italia S.r.l. Chimpanzee adenovirus vaccine carriers
US9951351B2 (en) 2014-10-09 2018-04-24 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
WO2023196550A3 (en) * 2022-04-07 2024-03-21 Regents Of The University Of Minnesota Adenovirus constructs and methods of use

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE278794T1 (en) * 1995-06-15 2004-10-15 Crucell Holland Bv PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRUSES, FOR USE IN GENE THERAPY
US6783980B2 (en) * 1995-06-15 2004-08-31 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US20040043489A1 (en) * 1998-07-08 2004-03-04 Menzo Havenga Gene delivery vectors provided with a tissue tropism for dendritic cells and methods of use
US6913922B1 (en) * 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US20050232900A1 (en) * 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
EP1157999A1 (en) * 2000-05-24 2001-11-28 Introgene B.V. Methods and means for enhancing skin transplantation using gene delivery vehicles having tropism for primary fibroblasts, as well as other uses thereof
US6579522B1 (en) * 2000-06-27 2003-06-17 Genvec, Inc. Replication deficient adenoviral TNF vector
CA2421864A1 (en) * 2000-09-20 2002-03-28 Crucell Holland B.V. Transduction of dendritic cells using adenoviral vectors
US7235233B2 (en) * 2000-09-26 2007-06-26 Crucell Holland B.V. Serotype 5 adenoviral vectors with chimeric fibers for gene delivery in skeletal muscle cells or myoblasts
US6821264B1 (en) 2000-11-27 2004-11-23 Gautam Khurana Gene delivery device and gene delivery method
EP1390476A4 (en) * 2001-04-17 2005-03-30 Vectorlogics Inc Mosaic adenoviral vectors
US20030219899A1 (en) * 2001-04-17 2003-11-27 Nikolay Korokhov Mosaic adenoviral vectors
US20040136963A1 (en) * 2001-06-22 2004-07-15 The Trustees Of The University Of Pennsylvania Simian adenovirus vectors and methods of use
US6905678B2 (en) * 2001-07-07 2005-06-14 Crucell Holland B.V. Gene delivery vectors with cell type specificity for mesenchymal stem cells
ATE467680T1 (en) * 2001-10-11 2010-05-15 Merck Sharp & Dohme HEPATITIS C VIRUS VACCINE
EP2301582B1 (en) * 2001-11-21 2017-07-12 The Trustees Of The University Of Pennsylvania Simian adenovirus nucleic acid and amino acid sequences, vectors containing same, and methods of use
AU2006204656B2 (en) * 2001-11-21 2008-01-24 The Trustees Of The University Of Pennsylvania Simian adenovirus nucleic acid and amino acid sequences, vectors containing same, and methods of use
EP1516055A4 (en) * 2002-01-24 2007-08-08 Scripps Research Inst Fiber shaft modifications for efficient targeting
AU2003241268A1 (en) * 2002-02-05 2003-09-09 Cornell Research Foundation, Inc. Adenoviral vector with replication-dependent transgene expression
PT1497438E (en) * 2002-04-25 2010-02-04 Crucell Holland Bv Means and methods for the production of adenovirus vectors
US20070275915A1 (en) * 2003-04-15 2007-11-29 Cell Genesys, Inc. Tmprss2 Regulatory Sequences and Uses Thereof
WO2004092396A2 (en) * 2003-04-15 2004-10-28 Novartis Ag Flap endonuclease 1 (fen1) regulatory sequences and uses thereof
US7291498B2 (en) * 2003-06-20 2007-11-06 The Trustees Of The University Of Pennsylvania Methods of generating chimeric adenoviruses and uses for such chimeric adenoviruses
US7491508B2 (en) * 2003-06-20 2009-02-17 The Trustees Of The University Of Pennsylvania Methods of generating chimeric adenoviruses and uses for such chimeric adenoviruses
ES2391975T3 (en) * 2003-07-25 2012-12-03 Genvec, Inc. Adenoviral vector based vaccines
US20070036721A1 (en) * 2003-09-23 2007-02-15 Uab Resarch Foundation Methods and compositions for in vivo inflammation monitoring
US7473418B2 (en) * 2004-03-25 2009-01-06 Cell Genesys, Inc. Pan cancer oncolytic vectors and methods of use thereof
WO2006008468A2 (en) * 2004-07-20 2006-01-26 The University Of Warwick Adenovirus vector and method to manipulate the adenovirus genome
US20060062764A1 (en) * 2004-08-25 2006-03-23 Seshidar-Reddy Police Fiber-modified adenoviral vectors for enhanced transduction of tumor cells
CA2589602A1 (en) * 2004-09-01 2006-04-13 Genvec, Inc. Adenoviral vectors able to transduce apcs, potential use in immune response generation
JP4843613B2 (en) * 2004-10-13 2011-12-21 クルセル ホランド ベー ヴェー Improved adenoviral vectors and methods of use thereof
US20060205080A1 (en) * 2005-03-01 2006-09-14 David Frey Formulations for therapeutic viruses having enhanced storage stability
US20060234347A1 (en) * 2005-04-13 2006-10-19 Harding Thomas C Targeting multiple angiogenic pathways for cancer therapy using soluble tyrosine kinase receptors
US9651543B2 (en) 2005-08-31 2017-05-16 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
US8450055B2 (en) * 2005-08-31 2013-05-28 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
EP2013348A4 (en) 2006-03-30 2009-09-02 Univ California Methods and compositions for localized secretion of anti-ctla-4 antibodies
US7919079B2 (en) * 2006-03-31 2011-04-05 Biosante Pharmaceuticals, Inc. Cancer immunotherapy compositions and methods of use
RS51250B (en) * 2006-04-28 2010-12-31 The Trustees Of The University Of Pennsylvania Modified adenovirus hexon protein and uses thereof
CA2675022A1 (en) * 2007-01-09 2008-07-17 Genvec, Inc. Adenoviral vector-based malaria vaccines
WO2011014794A1 (en) * 2009-07-31 2011-02-03 Paxvax, Inc. Adenoviral-based vectors
US9605844B2 (en) * 2009-09-01 2017-03-28 Cree, Inc. Lighting device with heat dissipation elements
EP2563387B1 (en) * 2010-04-28 2014-02-26 Institut Gustave Roussy Medical use of adenovirus vaccine vectors
SI2839014T1 (en) 2012-04-18 2021-05-31 The Children's Hospital Of Philadelphia Composition and methods for highly efficient gene transfer using aav capsid variants
GB2510321B (en) * 2012-11-12 2018-01-31 Biosurgical S L Agitation apparatus
WO2014082042A2 (en) * 2012-11-25 2014-05-30 The Regents Of The University Of California Peptides that stimulate subcutaneous adipogenesis
JP6576326B2 (en) 2013-03-14 2019-09-18 ソーク インスティテュート フォー バイオロジカル スタディーズ Oncolytic adenovirus composition
US10844102B2 (en) 2014-05-28 2020-11-24 The Regents Of The University Of California Peptides, compositions, and methods for stimulating subcutaneous adipogenesis
EP3292142B1 (en) * 2015-05-04 2024-03-20 Theriva Biologics S.L. Oncolytic adenoviruses with mutations in immunodominant adenovirus epitopes and their use in cancer treatment
EP3390428B1 (en) 2016-02-23 2019-09-25 Salk Institute for Biological Studies High throughput assay for measuring adenovirus replication kinetics
EP4155411A1 (en) 2016-02-23 2023-03-29 Salk Institute for Biological Studies Exogenous gene expression in therapeutic adenovirus for minimal impact on viral kinetics
CA3045892A1 (en) 2016-12-12 2018-06-21 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
CN111295449B (en) * 2017-10-31 2023-12-05 扬森疫苗与预防公司 Adenovirus vector and use thereof
MX2021005980A (en) 2018-11-21 2021-08-11 Mayo Found Medical Education & Res Adenoviruses and methods for using adenoviruses.
WO2020172509A1 (en) 2019-02-21 2020-08-27 Unleash Immuno Oncolytics, Inc. Oncolytic adenoviral vector and methods of use
EP3911671A1 (en) 2019-04-29 2021-11-24 Mayo Foundation for Medical Education and Research Multivalent pd-l1 binding compounds for treating cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996026281A1 (en) * 1995-02-21 1996-08-29 Genvec, Inc. Chimeric adenoviral fiber protein and methods of using same

Family Cites Families (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4593002A (en) * 1982-01-11 1986-06-03 Salk Institute Biotechnology/Industrial Associates, Inc. Viruses with recombinant surface proteins
US4487829A (en) * 1982-03-23 1984-12-11 Massachusetts Institute Of Technology Production and use of monoclonal antibodies against adenoviruses
US4589881A (en) * 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
US4517686A (en) * 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4797368A (en) * 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
FR2602790B1 (en) * 1986-08-13 1990-06-01 Transgene Sa EXPRESSION OF A SPECIFIC TUMOR ANTIGEN BY A RECOMBINANT VECTOR VIRUS AND USE THEREOF FOR THE PREVENTIVE OR CURATIVE TREATMENT OF THE CORRESPONDING TUMOR
EP0273085A1 (en) * 1986-12-29 1988-07-06 IntraCel Corporation A method for internalizing nucleic acids into eukaryotic cells
US5166320A (en) * 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US4956281A (en) * 1987-06-03 1990-09-11 Biogen, Inc. DNA sequences, recombinant DNA molecules and processes for producing lymphocyte function associated antigen-3
US5024939A (en) * 1987-07-09 1991-06-18 Genentech, Inc. Transient expression system for producing recombinant protein
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
JP2646013B2 (en) * 1988-09-14 1997-08-25 株式会社ジャパンエナジー Antiviral agent
US5204445A (en) * 1988-10-03 1993-04-20 The Scripps Research Institute Peptides and antibodies that inhibit integrin-ligand binding
US5198346A (en) * 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5096815A (en) * 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
EP0466815A4 (en) * 1989-04-05 1992-09-02 Novacell Corpporation Infectious targetted replication-defective virion
US5223394A (en) * 1989-04-10 1993-06-29 Biogen, Inc. Recombinant dna molecule comprising lymphocyte function-associated antigen 3 phosphatidylinositol linkage signal sequence
ATE144793T1 (en) * 1989-06-29 1996-11-15 Medarex Inc BISPECIFIC REAGENTS FOR AIDS THERAPY
US5240846A (en) * 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5332567A (en) * 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
US5436146A (en) * 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
DE69033181T2 (en) * 1989-10-20 1999-10-21 Dartmouth College IGA RECEPTOR-SPECIFIC MONOCLONAL ANTIBODIES
CA2069960C (en) * 1989-10-20 2004-03-02 Michael W. Fanger Bispecific heteroantibodies with dual effector functions
WO1991018088A1 (en) * 1990-05-23 1991-11-28 The United States Of America, Represented By The Secretary, United States Department Of Commerce Adeno-associated virus (aav)-based eucaryotic vectors
US5349053A (en) * 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
US5246921A (en) * 1990-06-26 1993-09-21 The Wistar Institute Of Anatomy And Biology Method for treating leukemias
GB2246779B (en) * 1990-08-03 1994-08-17 Delta Biotechnology Ltd Tumour-associated protease inhibitors targeted to tumour cells
AU660629B2 (en) * 1990-10-01 1995-07-06 University Of Connecticut, The Targeting viruses and cells for selective internalization by cells
GB9101550D0 (en) * 1991-01-24 1991-03-06 Mastico Robert A Antigen-presenting chimaeric protein
JP3534749B2 (en) * 1991-08-20 2004-06-07 アメリカ合衆国 Adenovirus-mediated gene transfer into the gastrointestinal tract
FR2681786A1 (en) * 1991-09-27 1993-04-02 Centre Nat Rech Scient RECOMBINANT VECTORS OF VIRAL ORIGIN, PROCESS FOR OBTAINING SAME AND THEIR USE FOR THE EXPRESSION OF POLYPEPTIDES IN MUSCLE CELLS.
IL103059A0 (en) * 1991-09-30 1993-02-21 Boehringer Ingelheim Int Conjugates for introducing nucleic acid into higher eucaryotic cells
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
SE503225C2 (en) * 1991-10-30 1996-04-22 Leif Lindholm Konsult Ab Virus-antibody complex for introduction of virus into mammalian cells
WO1993025234A1 (en) * 1992-06-08 1993-12-23 The Regents Of The University Of California Methods and compositions for targeting specific tissue
DE69333115D1 (en) * 1992-09-22 2003-08-28 Biofocus Discovery Ltd RECOMBINANT VIRUSES PRESENTING A NON-VIRAL POLYPEPTIDE ON THEIR OUTER SURFACE
AU692423B2 (en) * 1992-09-25 1998-06-11 Institut National De La Sante Et De La Recherche Medicale Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system, particularly in brain
GB9223084D0 (en) * 1992-11-04 1992-12-16 Imp Cancer Res Tech Compounds to target cells
EP0668913B1 (en) * 1992-11-18 2002-01-30 Arch Development Corporation Adenovirus-mediated gene transfer to cardiac and vascular smooth muscle
GB9300686D0 (en) * 1993-01-15 1993-03-03 Imp Cancer Res Tech Compounds for targeting
AU6133394A (en) * 1993-02-09 1994-08-29 Scripps Research Institute, The Targeting and delivery of genes and antiviral agents into cells by the adenovirus penton
DE4311651A1 (en) * 1993-04-08 1994-10-13 Boehringer Ingelheim Int Virus for the transport of foreign DNA into higher eukaryotic cells
ES2180579T3 (en) * 1993-05-10 2003-02-16 Univ Michigan TRANSFER OF GENES IN PANCREATIC EPITHELIAL CELLS.
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
EP0716711A1 (en) * 1993-09-03 1996-06-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Vector for gene therapy of the liver
US5552311A (en) * 1993-09-14 1996-09-03 University Of Alabama At Birmingham Research Foundation Purine nucleoside phosphorylase gene therapy for human malignancy
US5534423A (en) * 1993-10-08 1996-07-09 Regents Of The University Of Michigan Methods of increasing rates of infection by directing motion of vectors
FR2712812B1 (en) * 1993-11-23 1996-02-09 Centre Nat Rech Scient Composition for the production of therapeutic products in vivo.
IT1271461B (en) * 1993-12-01 1997-05-28 Menarini Ricerche Sud Spa ANTI-CD3 / ANTI-EGFR MONOCLONAL ANTI-BODY ANTIBODY, PROCESS FOR PRODUCTION AND ITS USE.
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5928944A (en) * 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US6312699B1 (en) * 1994-03-28 2001-11-06 Uab Research Foundation Ligands added to adenovirus fiber
US7252989B1 (en) * 1994-04-04 2007-08-07 Board Of Regents, The University Of Texas System Adenovirus supervector system
DE69520496T2 (en) * 1994-05-13 2001-07-12 Chiron Corp METHOD AND COMPOSITIONS AS VEHICLES FOR TARGETED INTRODUCTION OF GENES
US5571531A (en) * 1994-05-18 1996-11-05 Mcmaster University Microparticle delivery system with a functionalized silicone bonded to the matrix
FR2721943B1 (en) * 1994-06-29 1996-08-02 Rhone Poulenc Rorer Sa ADENOVIRUS COMPRISING A GENE ENCODING A SUPEROXIDE DISMUTASE
US5846782A (en) * 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
JPH10508743A (en) * 1994-09-09 1998-09-02 ニューロクライン バイオサイエンシーズ, インコーポレイテッド Interleukin-1 type 3 receptor
FR2724846B1 (en) * 1994-09-27 1996-12-20 Rhone Poulenc Rorer Sa METHOD OF TREATING CANCER BY REGULATING THE ACTIVITY OF RAS PROTEINS
JP3565859B2 (en) * 1994-10-28 2004-09-15 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア Improved adenovirus and uses thereof
WO1996014837A1 (en) * 1994-11-09 1996-05-23 Genetic Therapy, Inc. Gene therapy for hypercholesterolemia
ATE195555T1 (en) * 1994-11-29 2000-09-15 Takara Shuzo Co METHOD FOR PRODUCING TRANSFORMED CELLS
FR2727867B1 (en) * 1994-12-13 1997-01-31 Rhone Poulenc Rorer Sa GENE TRANSFER IN MEDULLAR MOTONURONES USING ADENOVIRAL VECTORS
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US5622699A (en) * 1995-09-11 1997-04-22 La Jolla Cancer Research Foundation Method of identifying molecules that home to a selected organ in vivo
WO1997024453A1 (en) * 1995-12-28 1997-07-10 Chiron Corporation Receptor specific chimeric viral surface polypeptides for viral and particle incorporation and internalization in target cells
WO1997038723A1 (en) * 1996-04-16 1997-10-23 Immusol Incorporated Targeted viral vectors
WO1998011221A2 (en) * 1996-09-13 1998-03-19 Dana-Farber Cancer Institute Car, a novel coxsackievirus and adenovirus receptor
CA2266342C (en) * 1996-09-25 2010-06-08 Novartis Ag Packaging cell lines for use in facilitating the development of high-capacity adenoviral vectors
US5877011A (en) * 1996-11-20 1999-03-02 Genzyme Corporation Chimeric adenoviral vectors
DE19649645A1 (en) * 1996-11-29 1998-06-04 Hoechst Ag Multiple functional ligand system for target cell-specific transfer of nucleotide sequences
US5922315A (en) * 1997-01-24 1999-07-13 Genetic Therapy, Inc. Adenoviruses having altered hexon proteins

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996026281A1 (en) * 1995-02-21 1996-08-29 Genvec, Inc. Chimeric adenoviral fiber protein and methods of using same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CRAWFORD-MIKSZA L AND SCHNURR P: "Analysis of 15 adenovirus hexon proteins reveal the location and structure of seven hypervariable regions containing serotype-specific residues", JOURNAL OF VIROLOGY, vol. 70, no. 3, March 1996 (1996-03-01), AMERICAN SOCIETY FOR MICROBIOLOGY US, pages 1836 - 1844, XP002071016 *
CROMPTON J ET AL.: "Expression of a forein epitope on the surface of the adenovirus hexon", JOURNAL OF GENERAL VIROLOGY, vol. 75, no. 1, January 1994 (1994-01-01), READING GB, pages 133 - 139, XP002071015 *
MASTRANGELI A ET AL.: ""Sero-switch" Adenovirus-mediated in vivo gene transfer: Circumvention of anti-adenovirus humoral immune defenses against repeat adenovirus vector administration by changing the adenovirus serotype", HUMAN GENE THERAPY, vol. 7, no. 1, 1 January 1996 (1996-01-01), pages 79 - 87, XP000653452 *

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6951755B2 (en) 1994-09-08 2005-10-04 Genvec, Inc. Vectors and methods for gene transfer
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US6576456B2 (en) 1995-02-21 2003-06-10 Cornell Research Foundation, Inc. Chimeric adenovirus fiber protein
US6057155A (en) * 1995-11-28 2000-05-02 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
EP0978566A2 (en) * 1998-07-08 2000-02-09 Introgene B.V. Chimaeric adenoviruses
WO2000003029A3 (en) * 1998-07-08 2000-03-16 Introgene Bv Chimaeric adenoviruses
EP0978566A3 (en) * 1998-07-08 2000-04-19 Introgene B.V. Chimaeric adenoviruses
WO2000003029A2 (en) * 1998-07-08 2000-01-20 Introgene B.V. Chimaeric adenoviruses
WO2000012738A1 (en) * 1998-08-27 2000-03-09 Aventis Pharma S.A. Targeted adenovirus vectors for delivery of heterologous genes
US6911199B2 (en) 1998-08-27 2005-06-28 Aventis Pharma S.A. Targeted adenovirus vectors for delivery of heterologous genes
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US6849446B2 (en) 2000-05-31 2005-02-01 University Of Saskatchewan Modified bovine adenovirus having altered tropism
US9228205B2 (en) 2000-11-15 2016-01-05 Crucell Holland B.V. Complementing cell lines
EP2824185A1 (en) 2003-02-24 2015-01-14 GenVec, Inc. Materials and methods for treating disorders of the ear
EP2168603A1 (en) 2003-11-14 2010-03-31 GenVec, Inc. Therapeutic regimen for treating cancer
EP3269390A3 (en) * 2004-01-23 2018-04-18 MSD Italia S.r.l. Chimpanzee adenovirus vaccine carriers
EP2567967A2 (en) 2004-04-12 2013-03-13 GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Method of using adenoviral vectors to induce an immune response
WO2006040330A2 (en) 2004-10-13 2006-04-20 Crucell Holland B.V. Improved adenoviral vectors and uses thereof
WO2006040330A3 (en) * 2004-10-13 2006-08-24 Crucell Holland Bv Improved adenoviral vectors and uses thereof
EA010433B1 (en) * 2004-10-13 2008-08-29 Круселл Холланд Б.В. Improved adenoviral vectors and uses thereof
US7741099B2 (en) 2004-10-13 2010-06-22 Beth Israel Deaconess Medical Center Inc. Adenoviral vectors and uses thereof
US8765146B2 (en) 2005-08-31 2014-07-01 Genvec, Inc. Adenoviral vector-based malaria vaccines
US8323663B2 (en) 2005-11-10 2012-12-04 Genvec, Inc. Adenoviral vector-based foot-and-mouth disease vaccine
WO2007148148A1 (en) * 2006-06-19 2007-12-27 Institut Gustave Roussy Inhibition of the liver tropism of adenoviral vectors
EP2385065A1 (en) 2007-11-01 2011-11-09 Perseid Therapeutics LLC Immunosuppressive polypeptides and nucleic acids
WO2009058564A2 (en) 2007-11-01 2009-05-07 Maxygen, Inc. Immunosuppressive polypeptides and nucleic acids
EP2612867A1 (en) 2007-11-01 2013-07-10 Perseid Therapeutics LLC Immunosuppressive polypeptides and nucleic acids
EP2612868A1 (en) 2007-11-01 2013-07-10 Perseid Therapeutics LLC Immunosuppressive polypeptides and nucleic acids
EP2325298A3 (en) * 2008-03-04 2012-08-29 The Trustees of The University of Pennsylvania Simian adenoviruses SAdV-36, -42.1, -42.2, AND -44 and uses thereof
WO2011047316A1 (en) 2009-10-16 2011-04-21 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Nucleic acid sequences encoding expandable hiv mosaic proteins
WO2011057248A2 (en) 2009-11-09 2011-05-12 Genvec, Inc. Simian adenovirus and methods of use
EP2853266A1 (en) 2009-11-09 2015-04-01 Genvec, Inc. Methods of propagating monkey adenoviral vectors
WO2011057254A2 (en) 2009-11-09 2011-05-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Simian adenoviral vector-based vaccines
EP3284821A1 (en) 2010-03-23 2018-02-21 Intrexon Corporation Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
US9493745B2 (en) 2010-04-14 2016-11-15 Mogam Biotechnology Research Institute Hexon isolated from simian adenovirus serotype 19, hypervariable region thereof and chimeric adenovirus using the same
EP2558481A4 (en) * 2010-04-14 2013-10-02 Mogam Biotech Res Inst Hexon isolated from simian adenovirus serotype 19, hypervariable region thereof and chimeric adenovirus using the same
EP2558481A1 (en) * 2010-04-14 2013-02-20 Mogam Biotechnology Research Institute Hexon isolated from simian adenovirus serotype 19, hypervariable region thereof and chimeric adenovirus using the same
US10066215B2 (en) 2010-04-14 2018-09-04 Mogam Biotechnology Research Institute Hexon isolated from simian adenovirus serotype 19, hypervariable region thereof and chimeric adenovirus using the same
WO2012021730A2 (en) 2010-08-11 2012-02-16 Genvec, Inc. Respiratory syncytial virus (rsv) vaccine
WO2012083297A2 (en) 2010-12-17 2012-06-21 Genvec, Inc. Adenoviral vectors with modified hexon regions
WO2012083297A3 (en) * 2010-12-17 2012-09-27 Genvec, Inc. Adenoviral vectors with modified hexon regions
WO2012088041A1 (en) 2010-12-20 2012-06-28 Genvec, Inc. Adenoviral vector-based dengue fever vaccine
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
EP3450568A2 (en) 2011-03-04 2019-03-06 Intrexon Corporation Vectors conditionally expressing protein
WO2013181128A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Modified serotype 28 adenoviral vectors
US9951351B2 (en) 2014-10-09 2018-04-24 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
US11279951B2 (en) 2014-10-09 2022-03-22 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
WO2018064523A1 (en) 2016-09-30 2018-04-05 Genvec, Inc. Adenovectors for delivery of therapeutic genetic material into t cells
WO2023196550A3 (en) * 2022-04-07 2024-03-21 Regents Of The University Of Minnesota Adenovirus constructs and methods of use

Also Published As

Publication number Publication date
AU6465298A (en) 1998-09-29
EP0973927A1 (en) 2000-01-26
CA2283628A1 (en) 1998-09-17
JP2001517080A (en) 2001-10-02
AU745252B2 (en) 2002-03-14
US6153435A (en) 2000-11-28
US6127525A (en) 2000-10-03

Similar Documents

Publication Publication Date Title
AU745252B2 (en) Chimeric adenoviral coat protein and methods of using same
US6455314B1 (en) Alternatively targeted adenovirus
US6649373B2 (en) Use of trans-activation and CIS-activation to modulate the persistence of expression of a transgene
ES2256302T3 (en) ADENOVIRAL VECTORS FOR THE TRANSDUCTION OF CONDROCITS.
US7968087B2 (en) Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
JP2000516098A (en) Short shaft adenovirus fiber and its use
US6849446B2 (en) Modified bovine adenovirus having altered tropism
US6867022B1 (en) Replication deficient adenovirus vectors and methods of making and using them
US9175309B2 (en) Recombinant adenovirus with enhanced therapeutic effect and pharmaceutical composition comprising said recombinant adenovirus
AU770780B2 (en) Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
EP1104813A1 (en) Conditional replication of recombinant human adenovirus DNA carrying modified inverted terminal repeat sequences

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 64652/98

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2283628

Country of ref document: CA

Ref country code: CA

Ref document number: 2283628

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1998 539879

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998910401

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1998910401

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 64652/98

Country of ref document: AU